Language selection

Search

Patent 3115039 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 3115039
(54) English Title: PDGF RECEPTOR ANTIBODY AND USE THEREOF
(54) French Title: ANTICORPS DE RECEPTEUR PDGF ET SON UTILISATION
Status: Examination Requested
Bibliographic Data
(51) International Patent Classification (IPC):
  • C07K 16/28 (2006.01)
  • A61K 47/68 (2017.01)
  • A61K 45/06 (2006.01)
  • A61P 27/02 (2006.01)
  • A61P 35/00 (2006.01)
  • C07K 16/44 (2006.01)
(72) Inventors :
  • KIM, SOOHYUN (Republic of Korea)
  • KIM, HYORI (Republic of Korea)
  • JO, DONG HYUN (Republic of Korea)
  • KIM, JEONG HUN (Republic of Korea)
  • KIM, SUREE (Republic of Korea)
  • KANG, DONGMIN (Republic of Korea)
  • HWANG, DOBEEN (Republic of Korea)
  • CHUNG, JUNHO (Republic of Korea)
(73) Owners :
  • SEOUL NATIONAL UNIVERSITY R&DB FOUNDATION (Republic of Korea)
  • EWHA UNIVERSITY-INDUSTRY COLLABORATION FOUNDATION (Republic of Korea)
The common representative is: SEOUL NATIONAL UNIVERSITY R&DB FOUNDATION
(71) Applicants :
  • SEOUL NATIONAL UNIVERSITY R&DB FOUNDATION (Republic of Korea)
  • EWHA UNIVERSITY-INDUSTRY COLLABORATION FOUNDATION (Republic of Korea)
(74) Agent: NORTON ROSE FULBRIGHT CANADA LLP/S.E.N.C.R.L., S.R.L.
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2019-10-07
(87) Open to Public Inspection: 2020-04-09
Examination requested: 2021-03-31
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/KR2019/013122
(87) International Publication Number: WO2020/071881
(85) National Entry: 2021-03-31

(30) Application Priority Data:
Application No. Country/Territory Date
62/741,733 United States of America 2018-10-05

Abstracts

English Abstract

The present invention relates to: a PDGF receptor antibody; an antibody-drug conjugate in which a chemotherapeutic agent is conjugated to the PDGF receptor antibody; and a use for preventing or treating angiogenic diseases by using same.


French Abstract

La présente invention concerne : un anticorps de récepteur PDGF ; un conjugué anticorps-médicament dans lequel un agent chimiothérapeutique est conjugué à l'anticorps de récepteur PDGF ; et une utilisation pour prévenir ou traiter des maladies angiogéniques à l'aide de celui-ci.

Claims

Note: Claims are shown in the official language in which they were submitted.


[CLAIMS]
[Claim 1]
An anti-PDGF receptor antibody or antigen-binding fragment thereof that
specifically
binds to platelet-derived growth factor receptor beta (PDGFR-f3) and comprises
a heavy chain
variable region and a light chain variable region,
wherein the heavy chain variable region of the anti-PDGF antibody receptor
comprises
VH-CDR1 comprising an amino acid sequence of SEQ ID NO: 1, 9, 17, 25, 33, 41,
or 49, VH-
CDR2 comprising an amino acid sequence of SEQ ID NO: 2, 10, 18, 26, 34, 42 or
50, and VH-
CDR3 comprising an amino acid sequence of SEQ ID NO: 3, 11, 19, 27, 35, 43 or
51, and
wherein the light chain variable region of the anti-PDGF receptor antibody
comprises
VL-CDR1 comprising an amino acid sequence of SEQ ID NO: 4, 12, 20, 28, 36, 44,
or 52, VL-
CDR2 comprising an amino acid sequence of SEQ ID NO: 5, 13, 21, 29, 37, 45 or
53, and VL-
CDR3 comprising an amino acid sequence of SEQ ID NOs: 6, 14, 22, 30, 38, 46 or
54.
[Claim 2]
The anti-PDGF receptor antibody or antigen-binding fragment thereof according
to
claim 1, wherein the anti -PDGF receptor antibody or antigen-binding fragment
thereof
specifically binds to an extracellular region of PDGFR-P.
[Claim 3]
The anti-PDGF receptor antibody or antigen-binding fragment thereof according
to
claim 1, wherein the anti-PDGF receptor antibody or antigen-binding fragment
thereof binds
non-competitively with PDGF-BB.
73

[Claim 4]
The anti-PDGF receptor antibody or antigen binding fragment thereof according
to
claim 3, wherein the anti-PDGF receptor antibody or antigen-binding fragment
thereof
comprises:
the heavy chain variable region is VH-CDR1 comprising an amino acid sequence
of
SEQ ID NO: 1, 33, 41 or 49, VH-CDR2 comprising an amino acid sequence of SEQ
ID NO:
2, 34, 42 or 50, and VH-CDR3 comprising an amino acid sequence SEQ ID NO: 3,
35, 43 or
51, and
the light chain variable region is VL-CDR1 comprising an amino acid sequence
of
SEQ ID NO: 4, 36, 44, or 52, VL-CDR2 comprising an amino acid sequence of SEQ
ID NO:
5, 37, 45 or 53, and VL-CDR3 comprising an amino acid sequence of SEQ ID NO:
6, 38, 46
or 54.
[Claim 5]
The anti-PDGF receptor antibody or antigen binding fragment thereof according
to
claim 1, wherein the anti-PDGF receptor antibody or antigen-binding fragment
thereof is
internalized by a cell expressing PDGFR-P.
[Claim 6]
The anti-PDGF receptor antibody or antigen binding fragment thereof according
to
claim 5, wherein the heavy chain variable region comprises VH-CDR1 comprising
an amino
acid sequence of SEQ ID NO: 1, 17, 25, 33, 41, or 49, VH-CDR2 comprising an
amino acid
sequence of SEQ ID NO: 2, 18, 26, 34, 42 or 50, and VH-CDR3 comprising an
amino acid
sequence of SEQ ID NO: 3, 19, 27, 35, 43 or 51, and
74

wherein the light chain variable region comprises VL-CDR1 comprising an amino
acid
sequence of SEQ ID NO: 4, 20, 28, 36, 44, or 52, VL-CDR2 comprising an amino
acid
sequence of SEQ ID NO: 5, 21, 29, 37, 45, or 53, and VL-CDR3 comprising an
amino acid
sequence of SEQ ID NO: 6, 22, 30, 38, 46 or 54.
[Claim 7]
The anti-PDGF receptor antibody or antigen-binding fragment thereof according
to
claim 1, wherein the anti-PDGF receptor antibody is an intact antibody in the
form of IgGl,
IgG2, IgG3 or IgG4.
[Claim 8]
The anti-PDGF receptor antibody or antigen binding fragment thereof according
to
claim 1, wherein the antigen-binding fragment of antibody is selected from the
group consisting
of scFv, (scFv)2, scFv-Fc, Fab, Fab' and F(ab')2.
[Claim 9]
The anti-PDGF receptor antibody or antigen-binding fragment thereof according
to
claim 8, wherein the antigen-binding fragment of antibody is a scFv comprising
a light chain
variable region (VL), a linker, and a heavy chain variable region (VH)
sequentially for N-
terminal to C-terminal direction.
[Claim 10]
The anti-PDGF receptor antibody or antigen-binding fragment thereof according
to
claim 8, wherein the anti-PDGF receptor antibody or antigen-binding fragment
thereof is

conjugated with a chemotherapeutic drug, a hapten, an enzyme, a peptide, an
aptamer, a toxin,
an affinity ligand, or a detection label.
[Claim 11]
The anti-PDGF receptor antibody or antigen-binding fragment thereof according
to
claim 10, wherein the hapten specifically binds to PDGFR-P.
[Claim 12]
The anti-PDGF receptor antibody or antigen-binding fragment thereof according
to
claim 10, wherein the hapten is cotinine, DNP (2,4-dinitrophenol), TNP (2,4,6-
trinitrophenol),
biotin, or digoxigenin.
[Claim 13]
The anti-PDGF receptor antibody or antigen-binding fragment thereof according
to
claim 10, wherein the hapten is bound to a chemotherapeutic drug.
[Claim 14]
A bispecific antibody, comprising the anti-PDGF receptor antibody or antigen-
binding
fragment thereof of any one of claims 1 to 9 which specifically binds to
platelet-derived growth
factor receptor beta (PDGFR-f3), and an antibody or antigen-binding fragment
thereof against
hapten for a chemotherapeutic drug.
[Claim 15]
The bispecific antibody according to claim 14, comprising an antigen-binding
fragment of an anti-PDGF receptor antibody, and an antigen-binding fragment of
an antibody
binding to a hapten.
76

[Claim 16]
The bispecific antibody according to claim 15, wherein a scFv of the anti-PDGF

receptor antibody and a scFv of the antibody against a hapten for a
chemotherapeutic drug are
connected directly or through a first linker.
[Claim 17]
The bispecific antibody of claim 16, wherein C-terminus of the scFv of the
anti-PDGF
receptor antibody and N-terminus of the scFv of the antibody against the
hapten for a
chemotherapeutic drug are connected through a first linker.
[Claim 18]
The bispecific antibody of claim 17, wherein the scFv of the anti-PDGF
receptor
antibody in which a heavy chain variable region and a light chain variable
region of the anti-
PDGF receptor antibody are connected through a second linker, and the scFv in
which a heavy
chain variable region and a light chain variable region of the anti-cotinine
antibody are
connected through a third linker are connected with the first linker,
wherein the heavy chain variable region of the anti-PDGF receptor antibody
comprises
VH-CDR1 comprising an amino acid sequence of SEQ ID NO: 1, 9, 17, 25, 33, 41,
or 49, VH-
CDR2 comprising an amino acid sequence of SEQ ID NO: 2, 10, 18, 26, 34, 42 or
50, and VH-
CDR3 comprising an amino acid sequence of SEQ ID NO: 3, 11, 19, 27, 35, 43 or
51, and
wherein the light chain variable region of the anti-PDGF antibody comprises VL-

CDR1 comprising an amino acid sequence of SEQ ID NO: 4, 12, 20, 28, 36, 44, or
52, VL-
CDR2 comprising an amino acid sequence of SEQ ID NO: 5, 13, 21, 29, 37, 45, or
53, and VL-
CDR3 comprising an amino acid sequence of SEQ ID NO: 6, 14, 22, 30, 38, 46 or
54.
77

[Claim 19]
The bispecific antibody of claim 14, wherein the chemotherapeutic drug is
duocarmycin, calicheamicin, pyrrolobenzodiazepine (PBD), anthracycline,
nemorubicin,
doxorubicin, Irinotecan, amatoxin, auristatin, maytansine, tubulysin, SN-38, 5-

Aminolaevulinic acid (ALA), Benzoporphyrin derivative monoacid ring A (BPD-
MA),
Chlorins, Tetra (m-hydroxyphenyl)chlorin (mTHPC), or Lutetium texaphyrin.
[Claim 20]
A drug delivery system for delivering a drug to cells expressing a PDGF
receptor,
comprising the anti-PDGF receptor antibody or antigen-binding fragment thereof
according to
any one of claims 1 to 9.
[Claim 21]
The drug delivery system according to claim 20, which the drug is an
immunotherapeutic agent or a chemotherapeutic agent.
[Claim 22]
The drug delivery system of claim 20, further comprising an antibody or
antigen-
binding fragment thereof against hapten for a chemotherapeutic drug, wherein
specifically
binds to the anti-PDGF receptor and the hapten.
[Claim 23]
A pharmaceutical composition for preventing, ameliorating or treating ocular
neovascular disease, comprising an anti-PDGF receptor antibody or antigen-
binding fragment
thereof according to any one of claims 1 to 9, or a bispecific antibody which
specifically binds
78

to PDGF receptor and happen and comprises the anti-PDGF receptor antibody or
antigen-
binding fragment thereof and an antibody or antigen-binding fragment thereof
against hapten
for a chemotherapeutic drug according to any one of claims 14 to 18; and drug.
[Claim 24]
The pharmaceutical composition according to claim 23, wherein the ocular
neovascular disease is ischemic retinopathy, iris neovascularization,
intraocular
neovascularization, senile Age-related macular degeneration, corneal
neovascularization,
retinal neovascularization, choroidal neovascularization, diabetic retinal
ischemia, or
proliferative diabetic retinopathy.
[Claim 25]
A pharmaceutical composition for preventing, ameliorating or treating cancer
disease,
comprising an anti-PDGF receptor antibody or antigen-binding fragment thereof
of any one of
claims 1 to 9, or a bispecific antibody which specifically binds to PDGF
receptor and happen
and comprises the anti-PDGF receptor antibody or antigen-binding fragment
thereof and an
antibody or antigen-binding fragment thereof against hapten for a
chemotherapeutic drug
according to any one of claims 14 to 18; and a chemotherapeutic drug.
79

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 03115039 2021-03-31
[Invention Title]
PDGF receptor antibody and use thereof
[Technical Field]
The present invention relates to an antibody against a PDGF receptor, an
antibody-
drug conjugate in which a chemotherapeutic agent is conjugated to the antibody
against a
PDGF receptor, and a prophylactic or therapeutic use for ocular neovascular
diseases using the
same.
[Related Art]
Angiogenesis, also called as neovascularization, involves the step of
formation of
protrusions from existing blood vessels and their infiltrating into
surrounding tissues.
Vasculogenesis, a process related to this, involves the differentiation of
endothelial cells and
hemangioblasts that already exist in the entire tissue, and then the formation
of blood vessels
as they connect together.
Angiogenesis occurs extensively during development, and also during wound
healing
to restore blood flow to tissues after injury or injury, even in a healthy
body. However,
angiogenesis is also associated with cancer and tumor formation.
Various ocular disorders involve alterations in angiogenesis. For example,
diabetic
retinopathy, one of the causes of blindness in adults, is associated with
excessive angiogenesis.
Non-proliferative retinopathy involves the selective loss of perivascular
cells in the retina, and
the associated capillaries expand and blood flow increases due to this loss.
In the expanded
capillary, endothelial cells proliferate and form cystic protrusions to become
microaneurysms,
and adjacent capillaries are blocked, so that perfusion does not occur in the
retinal region
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
around these microaneurysms. In fact, shunt blood vessels appear between
adjacent regions of
microaneurysms, and the clinical symptom of early diabetic retinopathy appears
to have
microaneurysms and non-perfused retinal regions. The leak occurs and
capillaries can burst in
microaneurysms, which can lead to effusion and bleeding. Proliferative
diabetic retinopathy
occurs when some areas of the retina continue to lose capillaries and become
unperfused,
resulting in new blood vessels appearing in discs and other areas on the
retina. These new blood
vessels easily grow into the vitreous and bleeding sites, causing preretinal
hemorrhage. As
proliferative diabetic retinopathy develops, excessive vitreous bleeding can
fill most of the
vitreous cavity. In addition, new blood vessels are accompanied by fibrous
tissue proliferation
that can cause traction retinal detachment.
Early treatment of macular edema and proliferative diabetic retinopathy
prevents
blindness for 5 years in 95% of patients, but delayed treatment prevents
blindness in only 50%
of patients. Therefore, early diagnosis and early treatment are essential.
Another ocular disorder accompanied with angiogenesis is senile macular
degeneration (AMD). AMD is characterized by a series of pathological changes
that occur in
the macula of the central region of the retina, and is accompanied by vision
problems that
specifically affect central vision. The retinal pigment epithelium resides on
Bruck's membrane,
which is a basement membrane complex being thickened and hardened in AMD. New
blood
vessels can be formed as they pass through Bruch's membrane from the lower
choroid which
contains an abundant layer of blood vessels. These blood vessels can then leak
fluid or cause
bleeding between the retinal pigment epithelium and the sensory retina, as
well as under the
retinal pigment epithelium. Subsequent fibrous scar formation causes loss of
central vision by
blocking the nutrients supply to photoreceptor cells and killing these cells.
This type of senile
2
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
maculopathy is called 'Vet type", because of leaking blood vessels and
subretinal edema or
blood. -Dry type" of senile maculopathy involves the collapse of the retinal
pigment epithelium
with the loss of the photoreceptor cells located over the retinal pigment
epithelium. This dry
type degrades vision.
Among these angiogenesis regulators, the role of PDGF-B members belonging to
the
PDGF family of signaling molecules is being studied, because they are believed
to play a
certain role in the formation, proliferation and proper functioning of
parietal cells such as
perivascular cells which are called as vascular smooth muscle or intervascular
cells.
The PDGF family consists of the monomers of PDGF-A, PDGF-B, PDGF-C and
PDGF-D and the dimers of PDGF-AA, PDGF-AB, PDGF-BB, PDGF-CC, and PDGF-DD.
Upon binding of the PDGF dimer, PDGF binds to the PDGF receptors a and (3. The
PDGF
receptor is a kind of tyrosine kinase enzyme, and can form a three combination
of aa, (3(3 and
c43 dimers. The extracellular region of the PDGF receptor is composed of five
immunoglobulin-
like regions, and the intracellular region contains a tyrosine phosphorylation
region. The ligand
binding site of the PDGF receptor is located in the first three immunoglobulin-
like regions.
PDGF binds to the immunoglobulin-like domains 2 and 3 of the PDGF receptor to
induce
dimerization of the receptor, and induces the direct receptor-receptor
interactions including
immunoglobulin-like domains 4 and 5 for further stabilization.
PDGF-CC specifically interacts with PDGF receptor-aa and PDGF receptor-4, but
does not interact with PDGFR-1313. Thus, it is similar to PDGF-AB. PDGF-DD is
considered as
being specific for PDGFR-(3(3, because it binds to PDGFR-(3(3 with high
affinity and to PDGF
receptor-4 with much lower degree. PDGF-AA only binds to the PDGF receptor-aa.
PDGF-
BB uniquely binds all three combinations of receptors with high affinity.
3
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
Excessive signaling between PDGF and PDGF receptor plays an important role in
many cancers such as renal cell carcinoma, lung cancer, glioblastoma, chronic
lymphocytic
leukemia, and prostate cancer. The PDGF receptor promotes tumor growth by
promoting tumor
persistence by directly affecting tumor growth related blood vessels, stromal
fibroblasts
(somatic fibroblasts) and the like.
As a monoclonal antibody targeting the signaling of PDGF and PDGF receptor,
Olaratumab (IMC-3G3, LartruvoTM) is a recombinant human IgG1 antibody against
PDGFR-
a that specifically binds to PDGF receptor a, blocks the binding of PDGF-AA,
PDGF-BB and
PDGF-CC and activation of the receptor, and dose not cross-react with PDGFR-
f3. Olaratumab
was first approved for cancer treatment in the U.S.A. in October 2016, based
on the results of
a stage lb/II studies of late soft tissue sarcoma that showed that the
combination of Olaratumab
and doxorubicin significantly improved overall mean survival compared to
doxorubicin alone.
Antibody-drug conjugates (ADCs) are therapeutic agents containing cytotoxic
drugs-
monoclonal antibodies, and delivering a toxic agent to cancer cells expressing
target antigens
with minimizing off-target toxicity.
In order to develop an optimized antibody-drug conjugate, there are
complicated
considerations such as antibody, linker, binding site, tumor type, antigen
expression rate, toxin,
drug to antibody ratio (DAR) and the like. For example, the binding site
affects the binding
rate, affinity, and stability. Because drug-conjugates internalize into cells
and release drugs,
toxic effects are important in how effectively they deliver drugs into cells.
In addition, the
internalization efficiency is different between antibodies that attach to the
same target antigen
located on the cell surface. Thus, the selection of an antibody internalizing
efficiently is very
important to the efficacy of the antibody-drug complex. Because there are many
factors, such
4
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
as the recycling of the antibody-drug conjugate to the target antigen, and
intracellular
trafficking affecting the cytotoxic effect of drugs, the method of accurately
measures the
toxicity rate of internalized antibodies can be checked after making the
antibody-drug
conjugate. Unfortunately, a lot of effort and time has been spent making
candidates of antibody-
drug conjugates using different types of antibodies.
Currently available PDGFR-P therapeutic agents focus on the antagonistic
effect of
PDGF ligand and cannot provide an efficient internalization of PDGFR-P.
Accordingly, there
is a need to develop an antibody that can efficiently internalize PDGFR-P to
deliver an antibody
or antibody-drug conjugate into cells expressing PDGFR-P.
[Disclosure]
[Technical Problem]
An embodiment of the present invention relates to an anti-PDGF receptor
antibody or
an antibody or its antigen-binding fragment that recognizes PDGF receptor,
specifically PDGF
receptor-P.
A further embodiment of the present invention relates to an antibody-drug
conjugate
in which a drug is conjugated to the anti-PDGF receptor antibody or antigen-
binding fragment
of the antibody. The antibody-drug conjugate may be a conjugate in which a
drug is bound to
an anti-PDGF receptor antibody through a non-covalent bond or a covalent bond.
Another embodiment of the present invention is a bispecific antibody
comprising the
anti-PDGF receptor antibody or antigen-binding fragment thereof, and an
antibody or antigen-
binding fragment thereof specifically recognizing a hapten capable of
conjugating with a drug.
The antigen-binding fragment of the antibody refers to a fragment of the
antibody
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
having at least one CDR, and may be scFv, (scFv)2, scFv-Fc, Fab, Fab' or
F(ab')2.
An embodiment of the present invention relates to a use of a drug delivery
system for
delivering a drug to cells or tissues expressing a PDGF receptor, specifically
PDGF receptor-13
on the cell surface, and comprising an anti-PDGF receptor antibody. The drug
delivery system
may be a bispecific antibody comprising an anti-PDGF receptor antibody or
antigen-binding
fragment thereof and an antibody or antigen-binding fragment thereof against
hapten for a
chemotherapeutic drug. When the drug delivery system includes the antibody or
antigen-
binding fragment thereof recognizing hapten, the drug may be connected to
hapten which is
connected to the anti-PDGF receptor antibody or an antigen binding fragment
thereof, and be
delivered to target cells or tissues. The drug delivered to the target cells
may be a drug for
preventing, ameliorating or treating diseases associated with PDGF or PDGF
receptor such as
neovascular disease. The drug may be an immunotherapeutic agent or a
chemotherapeutic
agent, and the immunotherapeutic agent may be an antibody.
An embodiment of the present invention relates to a pharmaceutical composition
for
preventing, ameliorating or treating neovascular diseases, comprising an
antibody-drug
conjugate in which a drug is conjugated to the anti-PDGF receptor antibody or
antigen-binding
fragment thereof, or an antibody-drug conjugate in which a drug is conjugated
via hapten to
the anti-PDGF receptor antibody or antigen-binding fragment thereof.
An embodiment of the present invention is a method of treating a subject
diagnosed as
suffering from a neovascular disease or having a risk of occurrence of a
neovascular disease,
comprising administering an antibody-drug conjugate in which a drug is
conjugated to the anti-
PDGF receptor antibody or antigen-binding fragment thereof, or an antibody-
drug conjugate
in which a drug is conjugated via hapten to the anti-PDGF receptor antibody or
antigen-binding
6
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
fragment thereof, to the subject at a sufficient amount of preventing,
improving or treating the
diseases in the subject as a first therapeutic agent or an auxiliary
therapeutic agent.
[Technical Solution]
Hereinafter, the present invention will be described in more detail.
An embodiment of the present invention relates to an antibody or antigen-
binding
fragment of an antibody that specifically recognizes a PDGF receptor,
specifically, PDGF
receptor-13 (PDGFR-13).
-Antibody" as used in the present disclosure is a substance produced through
antigen
stimulation. Examples of antibodies include, but are not limited to, animal
antibodies, chimeric
antibodies, humanized antibodies, and the like.
In addition, the isolated antigen-binding fragment is also within the scope of
the
antibody of the present invention. The complementary-determining region (CDR)
refers to a
variable region of an antibody that is important for antigen specificity. The
antigen-binding
fragment described above refers to a fragment of an antibody having at least
one CDR, which
may be scFv, (scFv)2, scFv-Fc, Fab, Fab' and F(ab')2, or preferably scFv. The
antibody may be
a polyclonal antibody or a monoclonal antibody. The antibody may be selected
from
immunoglobulins of all subtypes (e.g., IgA, IgD, IgE, IgG (IgGl, IgG2, IgG3,
IgG4), IgM,
etc.). The IgG type antibody may be an IgGl, IgG2, IgG3, or IgG4 subtype, such
as an IgG1
or IgG2 subtype.
As used in the present invention, the -antigen-binding fragment" of the chain
(heavy
chain or light chain) of an antibody or immunoglobulin is devoid of some amino
acids
compared to the full-length chain, but contains a portion of an antibody
capable of specifically
binding to an antigen. These antigen-binding fragments can be biologically
active in that they
7
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
can specifically bind to a target antigen or compete with other antibodies or
antigen-binding
fragments in order to bind to a specific epitope. Specifically, the antigen-
binding fragment is
an antibody fragment comprising one or more of the complementarity determining
regions, and
for example may be one selected from the group consisting of scFv, (scFv)2,
scFv-Fc, Fab, Fab'
and F(ab')2, but is not limited thereto. These biologically active fragments
can be produced by
recombinant DNA technology or, for example, by enzymatic or chemical cleavage
of intact
antibodies. Immunologically functional fragments of immunoglobulin are not
limited thereto.
Humanized" forms of non-human (e.g., chicken, mouse, rat, etc.) antibodies are

specific chimeric immunoglobulins, immunoglobulin chains or fragments thereof
that contain
minimal sequences derived from non-human immunoglobulins (e.g., For example,
Fv, Fab,
Fab', F(ab)2 or other antigen-binding products of the antibody). Generally, a
humanized
antibody is a human immunoglobulin (recipient antibody) in which the residues
obtained from
CDR of the recipient antibody is replaced with the residues having desired
specificity, affinity
and ability from CDRs of the non-human species antibody (donor antibody) such
as mice, rat
or rabbit. In some instances, Fv framework region (FR) residues of human
immunoglobulins
are replaced with corresponding non-human FR residues. In addition, the
humanized antibody
may contain residues which are not found in the CDR or FR sequences introduced
to the
recipient antibody. These modifications can be made to further refine and
optimize antibody
performance. In general, all or substantially all of the CDR regions in
humanized antibodies
correspond to regions of a non-human immunoglobulin, and all or substantially
all of the FR
residues may include at least entire of one, commonly two of variable domains
residues of a
human immunoglobulin consensus sequence. The humanized antibody will also
optimally
include at least a portion of an immunoglobulin constant region (Fc),
typically at least a portion
8
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
of a human immunoglobulin.
The present invention relates to an antibody against an extracellular domain
of the
PDGFR-13 isoform. PDGF receptor is a kind of tyrosine kinase enzyme, and
includes two
isoforms of PDGFR-a and PDGFR-13 which can form three types of combinations of
aa, f3f3
and c43 dimers. The structure of the PDGF receptor includes an extracellular
region composed
of five immunoglobulin-like domains, a transmembrane region, and an
intracellular region
(tyrosine kinase region). The ligand binding region of the receptor is located
in the front three
immunoglobulin-like regions among the five immunoglobulin-like domains of the
extracellular
region. PDGF is known to induce dimerization of the receptors by binding to
immunoglobulin-
like domains 2 and 3 of the PDGF receptor, and to direct receptor-receptor
interaction including
immunoglobulin-like domains 4 and 5 for additional stabilization.
The antibodies according to the invention may be human antibodies and antibody

fragments produced from the gene repertoire of immunoglobulin variable (V)
domains from
non-immunized donors in vitro, by using phage display technology (McCafferty
et al., (1990)
Nature, 348: 552-553).
The present invention relates to an isolated antibody or antigen-binding
fragment
thereof that specifically binds to PDGFR-13, wherein the antibody or antigen-
binding fragment
may be a polypeptide, protein or antibody or an antigen-binding fragment
thereof including a
complementarity determining region of a heavy chain and a complementarity
determining
region of a light chain, to specifically bind to PDGFR-f3.
In a specific example, the anti-PDGFR-13 antibody and antigen-binding fragment

thereof may include:
9
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
(i) at least one heavy chain complementarity determining region selected from
the
group consisting of H-CDR1, H-CDR2 and H-CDR3, or a heavy chain variable
region
including the at least one heavy chain complementarity determining region;
(ii) at least one light chain complementarity determining region selected from
the
group consisting of L-CDR1, L-CDR2, and L-CDR3, or a light chain variable
region including
the at least one light chain complementarity determining region;
a combination of the at least one heavy chain complementarity determining
region and
the at least one light chain complementarity determining region; or
a combination of the heavy chain variable region and the light chain variable
region.
Additionally, in the heavy chain variable region, the light chain variable
region, or a
combination of the heavy chain variable region and the light chain variable
region, the heavy
chain variable region may include one or more heavy chain frameworks selected
from the group
consisting of H-FR1, H-FR2, H-FR3 and H-FR4, and the light chain variable
region may
include one or more light chain frameworks selected from the group consisting
of L-FR1, L-
FR2, L-FR3, and L-FR4.
The heavy chain complementarity determining region of (i) H-CDR1, H-CDR2 or H-
CDR3 according to the present invention is selected from amino acid sequences
shown in
Tables 1 and 2 below, or incudes at least one amino acid sequence having
substantial sequence
identity with the selected amino acid sequence. In addition, (ii) the light
chain complementarity
determining region of L-CDR1, L-CDR2 or L-CDR3 is selected from amino acid
sequences
shown in Tables 1 and 2 below, or includes at least one amino acid sequence
having substantial
sequence identity with the selected amino acid sequence.
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
In the present invention, the single-chain Fv is a single polypeptide chain of
an antigen-
binding region in which the heavy variable region and light chain variable
region are connected
directly or by a linker, and can be at least one selected from scFv of the
heavy chain variable
region and the light chain variable region are linked in a single chain form,
scFv dimers (di-
scFv), and scFv-Fc in which a heavy chain variable region, a light chain
variable region, and
Fc are linked in a single chain form. The scFv may be the heavy chain variable
region and light
chain variable region connected by a linker that is not particularly limited,
and for example,
may be a linker sequence having an amino acid sequence of SEQ ID NO: 57 in
Table 2 below.
The present invention includes an antibody against a platelet-derived growth
factor
receptor beta (PDGFR-f3) of mouse or human as an example of target antigen,
and scFv of the
antibody can be used for producing a bispecific antibody. For example, the
scFv may be an
antibody in which a heavy chain having CDR1 to CDR3 and a light chain having
CDR1 to
CDR3 of an antibody are connected by a linker. Additionally, as a bispecific
antibody, the
fusion protein of (scFv of anti-PDGFR-13 antibody)-C-K-(scFv of anti-cotinine
antibody) may
include three(3) CDRs of the heavy chain and three(3) CDRs of the light chain
of each antibody
which can be linked to CI< as specified in Table 2 below.
In a specific example, specific examples of antibodies against mouse PDGFR-13
(mPDGFR-13) as an example of a target antigen according to the present
invention, are PRb-
CN01, PRb-CC01, PRb-CCO2 and PRb-CC03, and their heavy chain variable region,
light
chain variable region, and CDR sequences of these regions are shown in Table 1
below.
[Table 1]
Clone/name Part Amino acid sequence (N->C) SEQ ID
NO
PRb-CNO1 CDR1- GFTFSDRAMH 1
VH
11
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
PRb-CN01 CDR2- LITNTGGSTNYGAAVKG 2
VH
PRb-CN01 CDR3- GVGSWAHGGRIDA 3
VH
PRb-CN01 CDR1- SGGSGSYG 4
VL
PRb-CN01 CDR2- SNNQRPS 5
VL
PRb-CN01 CDR3- GTRD SSYVGI 6
VL
PRb-CN01 VH AVTLDESGGGLQTPGGALSLVCKASGFT 7
FSDRAMHWVRQAPGKGLEWVGLITNTG
GSTNYGAAVKGRATISRDNGQ STVRLQ
LNNLRAEDTGTYYCTRGVGSWAHGGRI
DAWGHGTEVIVSS
PRb-CNO1 VL LT QP S SVSANP GETVKITC SGGSGSYGW 8
FQ QKSPGSAPVTVIYSNNQRP SD IP SRF S
GSKSGSTNTLTITGVQAEDEAIYYCGTR
DSSYVGIFGAGTTLTVL
PRb-CC 01 CDR1- SGFTFS SYNMG 9
VH
PRb-CC 01 CDR2- GI SAADNSTAYGAAVDG 10
VH
PRb-CC 01 CDR3- GGGS IDA 11
VH
PRb-CC 01 CDR1- SGGGSYYG 12
VL
PRb-CC 01 CDR2- YNDKRPS 13
VL
PRb-CC 01 CDR3- GAWDNSAGYAG 14
VL
PRb-CC 01 VH AVTLDESGGGLQTPGGALSLVCKGSGFT 15
FS SYNMGWVRQAPGKGLEFVAGISAAD
NSTAYGAAVDGRATISRDNGQ STVRLQ
LNNLRAEDTATYFC IRGGGSIDAWGH GT
EVIVSS
PRb-CC 01 VL LT QP S SVSANLGGTVKITC SGGGSYYGW 16
YQQKSPGSAPVTLIYYNDKRPSDIP SRFS
GSKSGSTGTLTITGVQAEDEAVYFCGAW
DNSAGYAGFGAGTTLTVL
PRb-CC 02 CDR1- GFTFSSYGMF 17
VH
PRb-CC 02 CDR2- GIDT GS GTNYGSAVKG 18
VH
PRb-CC 02 CDR3- GYAGTIDA 19
VH
PRb-CC 02 CDR1- SGGGGSYG 20
VL
12
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
PRb-CCO2 CDR2- YNDKRPS 21
VL
PRb-CCO2 CDR3- GSYNSSDSL 22
VL
PRb-CCO2 VH
AVTLDESGGGLQTPGGALSLVCKASGFT 23
FS SYGMFWVRQAPGKGLEWVAGIDTGS
GTNYGSAVKGRATISRDNGQSTVRLQL
NNLRAEDTGTYFCTRGYAGTIDAWGHG
TEVIVSSTS
PRb-CCO2 VL
LTQPSSVSANPGETVKITCSGGGGSYGW 24
YQQKSPGSAPVTVIYYNDKRPSDIPSRFS
GSKSGSTGTLTITGVQAEDEAVYFCGSY
NSSDSLFGAGTTLTVL
PRb-CCO3 CDR1- GFTFSSYAMG 25
VH
PRb-CCO3 CDR2- GIDTAGGTAYGPAVKG 26
VH
PRb-CCO3 CDR3- SSYIDT 27
VH
PRb-CCO3 CDR1- SGGTYNYG 28
VL
PRb-CCO3 CDR2- WNDKRPS 29
VL
PRb-CCO3 CDR3- GSSDSSGLI 30
VL
PRb-CCO3 VH
AVTLDESGGGLQTPGGTLSLVCKASGFT 31
FS SYAMGWVRQAPGKGLEWVAGIDTA
GGTAYGPAVKGRATISRDNGQSTVRLQ
LNNLRAEDTATYYCTRSSYIDTWGHGTE
VIVSSTS
PRb-CCO3 VL
LTQPSSVSANPGETVKITCSGGTYNYGW 32
YQQKSPGSAPVTVIYWNDKRPSDIPSRFS
GSKSGSTGTLTITGVQAEDEAVYYCGSS
DSSGLIFGAGTTLTVL
Among PRb-CN01, PRb-CC01, PRb-CCO2 and PRb-CCO3 antibodies, PRb-CC01,
PRb-CCO2, and PRb-CCO3 compete with mPDGF-BB, and the binding site is
immunoglobulin-like domain 2 or 3 of PDGFR-13, but PRb-CN01 does not compete
with
mPDGF-BB and is expected to bind to the immunoglobulin-like domain 1, 4 or 5
of PDGFR-
(3. It was confirmed that both PRb-CN01 and PRb-32 showed the equivalent
cytotoxicity
13
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
regardless of the presence or absence of mPDGF-BB and that PRb-CNO1 and PRb-32
were
surrogate antibodies (Fig. 20a and Fig. 20b).
The novel antibodies according to the present invention, such as PRb-CNO1, PRb-

CC01, PRb-CCO2 and PRb-CCO3 antibodies, antibody fragments thereof, mixtures
thereof or
derivatives thereof have binding affinity in the range of 1 x 10-7M to 1 x 10-
10 M for the PDGF
receptor, when they are used for producing a bispecific antibody with an anti -
cotinine antibody.
It was confirmed that the PRb-CNO1, PRb-CCO2 and PRb-CCO3 antibodies
internalized into cells through endosomes, but PRb-CCO1 was not internalized.
Because
antibody-drug conjugates internalize into cells and release drugs, toxic
effects are important in
how effectively they deliver drugs into cells. In addition, the
internalization efficiency is
different between antibodies that attach to the same target antigen on the
cell surface. Thus, the
selection of an antibody internalizing efficiently is very important in the
efficacy of the
antibody-drug complex.
In addition, the PRb-CNO1, PRb-CC01, PRb-CCO2 and PRb-CCO3 antibodies exhibit
cytotoxicity, and PRb-CNO1 among the four antibodies has the highest
cytotoxicity when it
forms a complex with cotinine-duocarmycin. Therefore, it can be considered as
the most
effective targeting vehicle for drugs such as duocarmycin. Even after adding
mPDGF-BB, the
cytotoxicity of the bispecific including PRb-CNO1 complexed with cot-duo or
cot-duo-cot was
still high. However, when mPDGF-BB was added, the toxicity of the three
antibodies (PRb-
CC01, PRb-CCO2 and PRb-CCO3) complexed with cot-duo or cot-duo-cot competing
with
mPDGF-BB was slightly reduced. In the present invention, the bispecific
antibody related with
PRb-CNO1 did not compete with PDGF-BB and induced cytotoxicity regardless of
the
presence of PDGF-BB (FIGS. 5a to 5b).
14
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
Specific examples of antibodies against human PDGFR-13 (hPDGFR-f3) which are
examples of target antigens according to the present invention are PRb-CN16,
PRb-CN32, and
PRb-CN26. The CDR sequences of these antibodies are shown in Table 2 below.
[Table 2]
Clone/name Part Sequence SEQ ID
NO
PRb-CN16 CDR1-VH GFTF S S FNMA 33
PRb-CN16 CDR2-VH EISNTAGSTFYAPAVKG 34
PRb-CN16 CDR3-VH AAGTCYSHSCTGYIDA 35
PRb-CN16 CDR1-VL SGSS SSYG 36
PRb-CN16 CDR2-VL ENNQRPS 37
PRb-CN16 CDR3-VL GNADRSNSAGT 38
PRb-CN16 VH
AVTLDESGGGLQTPGTALSLVCKASGF 39
TFS SFNMAWVRQAPGKGLEFVGEISNT
AGSTFYAPAVKGRATISRDNGQ STVRL
QLNNLRAEDTAIYYCAKAAGTCYSH SC
TGYIDAWGHGTEVIVS ST S
PRb-CN16 VL LT QP S SVSANP GETVKITC S GS SS SYGW 40
YQQKSPGSAPVTLIYENNQRPSNIPSRF
S GSKS GST GT LTIT GVQAEDEAVYYC G
NADRSNSAGTFGAGTTLTVL
PRb-CN32 CDR1-VH GFTFSSFNMF 41
PRb-CN32 CDR2-VH GISTTGRYTGYGSAVQG 42
PRb-CN32 CDR3-VH SAGSTYSYWDSDAGLIDA 43
PRb-CN32 CDR1-VL SGGSNAGSYYYG 44
PRb-CN32 CDR2-VL SNNQRPS 45
PRb-CN32 CDR3-VL GS GD SSSIAA 46
PRb-CN32 VH
AVTLDESGGGLQTPRGTLSLVCKASGF 47
TFS SFNMFWVRQAPGKGLEFVAGISTT
GRYTGYGSAVQGRGTISRDNGQ STVRL
QLNNLRAEDTGTYYCAKSAGSTYSYW
D SDAGLIDAWGH GTEVIVS ST S
PRb-CN32 VL LT QP S
SVSANLGETVKITC S GGSNAG SY 48
YYGWYQQKSPGSAPVTVIYSNNQRP S
DIPSRFSGSTSGSTSTLTITGVQVDDEA
VYFC GS GD S S SIAAFGAGTTLTVL
PRb-CN26 CDR1-VH GFTFSDRGIH 49
PRb-CN26 CDR2-VH GIGNTGSYTAYAPAVKG 50
PRb-CN26 CDR3-VH RGFMDAGGIDA 51
PRb-CN26 CDR1-VL SGGGSYAGSYYYG 52
PRb-CN26 CDR2-VL DNTNRPS 53
PRb-CN26 CDR3-VL GGYD GI SDTGI 54
PRb-CN26 VH
AVTLDESGGGLQTPGGGLSLVCKASGF 55
TFSDRGIHWVRQAPGKGLEWVAGIGN
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
TGSYTAYAPAVKGRATISRDNGQSTVR
LQLNNLRAEDTATYYCAKRGFMDAGG
IDAWGHGTEVIVSSTS
PRb-CN26 VL LTQPSSVSANPGETVKITCSGGGSYAGS 56
YYYGWYQQKSPGSAPVTVIYDNTNRP
SNIPSRFSGSLSGSTDTLTITGVQAEDEA
VYFCGGYDGISDTGIFGAGTTLTVL
Linker VL-VH GQSSRSSGGGGSSGGGGS 57
(for
producing
scFv)
Kappa * RTVAAPSVFIFPPSDEQLKSGTASVVCL 58
constant LNNFYPREAKVQWKVDNALQSGNSQE
(Ck) SVTEQDSKDSTYSLSSTLTLSKADYEK
HKVYACEVTHQGLSLPVTKSFNRGES
Linker scFv-Ck (GGGGS)3 59
(for
producing
bispecific
antibody)
The PRb-CN16, PRb-CN32, and PRb-CN26 antibodies do not compete with hPDGF-
BB, and thus, are expected to bind to immunoglobulin-like domains 1, 4 or 5 of
hPDGFR-13.
All of PRb-CN16, PRb-CN32 and PRb-CN26 antibodies are confirmed to bind to
hPDGFR-13,
mPDGFRP, Rhesus PDGFRP, Cynomolgus monkey PDGFRP, and Sus crowfa PDGFR-P. It
was confirmed that both PRb-CNO1 and PRb-32 showed the equivalent cytotoxicity
regardless
of the presence/absence of mPDGF-BB, and PRb-CNO1 and PRb-32 were surrogate
antibodies
(Fig. 20a and Fig. 20b).
The novel antibodies according to the present invention, such as PRb-CN16, PRb-

CN32, and PRb-CN26 antibodies, antibody fragments, mixtures thereof or
derivatives thereof,
have binding affinity 1 x 10-7M to 1 x 10-11 M to PDGF receptor, in the case
that they are used
for preparing a bispecific antibody with an anti -cotinine antibody.
In the evaluation of the cellular internalization performance of an antibody
against
human PDGFR-13 (hPDGFR-13) as an example of a target antigen according to the
present
16
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
invention in the absence of hPDGF-BB, PRb-CN32 internalizes most effectively
to human
pericyte cells, PRb-CN16 is the second performance, and PRb-CN26 is not
internalized. Also,
in the evaluation of the cellular internalization performance with treatment
by hPDGF-BB, all
of PRb-CN16, PRb-CN32, and PRb-CN26 antibodies have excellent cell
internalization
performance. When preparing an antibody-drug conjugate, it is internalized
into the cell and
releases drug. Therefore, the hPDGFR43 antibody according to the present
invention moves to
the target cell and internalizes well. When the antibody-drug conjugate is
prepared using the
antibody, it is internalized into the cell and releases the drug, resulting in
contributing to the
increased drug efficacy. Patients with diseases associated with problematic
wet type AMD and
PDGFR-f3 have increased expression level of PDGFR-13 and/or increased
expression level of
PDGF-BB, and these increased expression levels tend to inhibit internalization
or efficacy of
antibodies. In this regard, the anti-PDGFR-13 antibody according to the
present invention has
an advantage that the internalization level of the antibody is not suppressed
or the
internalization is further increased, even if the expression level of PDGF-BB
is increased.
When PRb-CN32 and PRb-CN26 form a complex with cotinine-duocarmycin among
the PRb-CN16, PRb-CN32 and PRb-CN26 antibodies, they exhibit the highest
cytotoxicity.
Therefore, they can be considered as most effective targeting vehicle for
drugs such as
duocarmycin.
All of PRb-CN16, PRb-CN32, and PRb-CN26 showed higher cytotoxicity than the
control anti-mCD154 x cotinine scFv-Cx-scFy in PDGFR-P-expressing cell lines
with or
without hPDGF-BB, and especially, PRb-CN26 and PRb-CN32 showed high
cytotoxicity (Figs.
14a and 14b). In addition, all of PRb-CN16, PRb-CN32, and PRb-CN26 have
equivalent
cytotoxicity in a cell line that does not express hPDGFR-13, compared to the
control anti-
17
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
mCD154 x cotinine scFv-C-K-scFv. Therefore, the bispecific antibody-drug
(IC50) has different
toxicity in cell lines not expressing hPDGFR-13 and cell lines expressing
hPDGFR-13 , shows
toxicity at a lower concentration in hPDGFR-13 expressing cell lines, and at
that concentration,
has higher toxicity in pathological cells or tissues expressing much hPDGFR-
13, with
minimizing toxicity in normal cells or normal tissues. In addition, patients
with diseases related
problematic Wet type AMD and PDGFR-13 have increased PDGFR-13 expression
levels (C.
Zehetner et al., Invest Ophthalmol Vis Sci. 55 (2014) 337-44) and/or increased
PDGF-BB
expression level (C. Zehetner, R. Kirchmair, SB Neururer, MT Kralinger, NE
Bechrakis, GF
Kieselbach, Systemic upregulation of PDGF-B in patients with neovascular AMD,
Invest
Ophthalmol Vis Sci. 55 (2014) 337-44)), and these increased expression levels
tend to inhibit
the internalization or efficacy of the antibody. In this regard, the anti-
PDGFR-13 antibody
according to the present invention has the advantage that the internalization
level of the
antibody is not suppressed or the internalization is further increased, even
if the expression
level of PDGF-BB is increased.
More specifically, the antibody according to the present invention
specifically binds
to platelet-derived growth factor receptor beta (PDGFR-13), and may be anti-
PDGF receptor
antibody or antigen binding fragment thereof including a heavy chain variable
region and a
light chain variable region,
wherein the heavy chain variable region of the anti -PDGF receptor antibody
includes
VH-CDR1 comprising an amino acid sequence of SEQ ID NO: 1, 9, 17, 25, 33, 41,
or 49, VH-
CDR2 comprising an amino acid sequence of SEQ ID NO: 2, 10, 18, 26, 34, 42 or
50, and VH-
CDR3 comprising an amino acid sequence of SEQ ID NO: 3, 11, 19, 27, 35, 43 or
51, and
18
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
wherein the light chain variable region of the anti-PDGF receptor antibody is
VL-
CDR1 comprising an amino acid sequence of SEQ ID NO: 4, 12, 20, 28, 36, 44, or
52, VL-
CDR2 comprising an amino acid sequence of SEQ ID NO: 5, 13, 21, 29, 37, 45, or
53, and VL-
CDR3 comprising an amino acid sequence of SEQ ID NOs: 6, 14, 22, 30, 38, 46 or
54.
The anti-PDGF receptor antibody or antigen-binding fragment thereof binds non-
competitively to PDGF-BB, and includes, for example, the heavy chain variable
region
includes VH-CDR1 comprising an amino acid sequence of SEQ ID NO: 1, 33, 41 or
49, VH-
CDR2 comprising an amino acid sequence of SEQ ID NO: 2, 34, 42 or 50, and VH-
CDR3
comprising an amino acid sequence of SEQ ID NO: 3, 35, 43 or 51, and
the light chain variable region including VL-CDR1 comprising an amino acid
sequence of SEQ ID NO: 4, 36, 44, or 52, VL-CDR2 comprising an amino acid
sequence of
SEQ ID NO: 5, 37, 45, or 53, VL-CDR3 comprising an amino acid sequence of SEQ
ID NO:
6, 38, 46 or 54.
The anti-PDGF receptor antibody or antigen-binding fragment thereof may be
internalized by a cell expressing PDGFR-13, and include, for example,
the heavy chain variable region includes VH-CDR1 comprising an amino acid
sequence of SEQ ID NO: 1, 17, 25, 33, 41, or 49, VH-CDR2 comprising an amino
acid
sequence of SEQ ID NO: 2, 18, 26, 34, 42 or 50, and VH-CDR3 comprising an
amino acid
sequence of SEQ ID NO: 3, 19, 27, 35, 43 or 51, and
the light chain variable region includes VL-CDR1 comprising an amino acid
sequence
of SEQ ID NO: 4,20, 28, 36, 44, or 52, VL-CDR2 comprising an amino acid
sequence of SEQ
ID NO: 5, 21, 29, 37, 45, or 53, and VL-CDR3 comprising an amino acid sequence
of SEQ ID
NO: 6, 22, 30, 38, 46 or 54.
19
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
Specific anti-PDGF receptor antibody or antigen-binding fragment thereof of
the
present invention may include:
(a) VH-CDR1 to VH-CDR3 each comprising an amino acid sequence of SEQ ID NOs:
1 to 3 and VL-CDR1 to VL-CDR3 each comprising an amino acid sequence of SEQ ID
NOs:
4 to 6,
(b) VH-CDR1 to VH-CDR3 each comprising an amino acid sequence of SEQ ID NOs:
9 to 11 and VL-CDR1 to VL-CDR3 each comprising an amino acid sequence of SEQ
ID NOs:
12 to 14,
(c) VH-CDR1 to VH-CDR3 each comprising an amino acid sequence of SEQ ID NOs:
17 to 19 and VL-CDR1 to VL-CDR3 each comprising an amino acid sequence of SEQ
ID NOs:
20 to 22,
(d) VH-CDR1 to VH-CDR3 each comprising an amino acid sequence of SEQ ID NOs:
25 to 27 and VL-CDR1 to VL-CDR3 each comprising an amino acid sequence of SEQ
ID NOs:
28 to 30,
(e) VH-CDR1 to VH-CDR3 each comprising an amino acid sequence of SEQ ID NOs:
33 to 35 and VL-CDR1 to VL-CDR3 each comprising an amino acid sequence of SEQ
ID NOs:
36 to 38,
(0 VH-CDR1 to VH-CDR3 each comprising an amino acid sequence of SEQ ID NOs:
41 to 43 and VL-CDR1 to VL-CDR3 each comprising an amino acid sequence of SEQ
ID NOs:
44 to 46, or
(g) VH-CDR1 to VH-CDR3 each comprising an amino acid sequence of SEQ ID
NOs: 49 to 50 and VL-CDR1 to VL-CDR3 each comprising an amino acid sequence of
SEQ
ID NOs: 52 to 54.
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
An embodiment of the present invention relates to a use of a drug delivery
system that
delivers a drug to cells or tissues having PDGF receptor, specifically PDGF
receptor-13 on a
cell surface, and includes an anti-PDGF receptor antibody. The anti-PDGF
receptor antibody
or antigen-binding fragment thereof is targeted to a target cell having PDGF
receptor and is
internalized to the inside of target cell, so as to efficiently deliver
substances conjugated to the
antibody or antigen-binding fragment thereof into the target cell. The drug to
be delivered is a
drug capable of binding to an anti-PDGF receptor antibody directly or through
a linker, so as
to form an antibody-drug conjugate, or a hapten-drug conjugate to bind to an
anti-PDGF
receptor antibody through a hapten.
An embodiment of the present invention provides a complex including an anti-
PDGF
receptor antibody or antigen-binding fragment thereof being capable of a
conjugating
compound such as a chemotherapeutic drug, enzyme, peptide, aptamer, toxin,
affinity ligand,
or detection label, and a conjugating compound. Depending on the type of the
conjugating
compound, the complex may be variously applied to diagnosis or treatment of a
disease.
A further embodiment of the present invention relates to an antibody-drug
conjugate
in which a drug is conjugated to the anti-PDGF receptor antibody or antigen-
binding fragment
of the antibody. The antibody-drug conjugate may be a conjugate in which a
drug is bound to
an anti-PDGF receptor antibody by a non-covalent bond or covalent bond.
As used herein, -conjugating body" or -conjugate" refers to an antibody or
antigen-
binding fragment thereof and other molecule disclosed hereafter, particularly
a chimeric
molecule of a therapeutic agent described below. In the conjugate, the
antibody or antigen-
binding fragment thereof according to the present invention is linked
physically to the other
molecule by covalent bond or physical force of Van Der Waals force or
hydrophobic interaction,
21
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
encapsulation, embedding or a combination of the above. In the conjugate
according to an
embodiment, the antibody or antigen-binding fragment thereof according to the
present
invention may be linked through a peptide linker.
Another embodiment of the present invention may be a bispecific antibody
including
an anti-PDGF receptor antibody or antigen-binding fragment thereof
specifically recognizing
hapten that is capable of conjugating with a drug. The antigen-binding
fragment of the antibody
refers to a fragment of an antibody having at least one CDR, and may be scFv,
(scFv)2, scFv-
Fc, Fab, Fab' and F(ab')2. In the present invention, -multispecific antigen-
binding protein" or
-multispecific antibody" targets two or more antigens or epitopes, and
includes two or more
antigen-binding sites. In the present invention, -bispecific" or -dual
specific" antigen binding
protein or antibody is a hybrid antigen binding protein or antibody having two
different antigen
binding sites. Such a bispecific antibody is a kind of multispecific antigen
binding protein or
multispecific antibody, and can be produced by various known methods, for
example, fusion
of hybridomas or connection of Fab' fragments.
In case that the drug delivery system according to the present invention
includes a
hapten-recognizing antibody or antigen-binding fragment thereof, the drug may
bind to hapten
and then binds to the anti-PDGF receptor antibody or antigen-binding fragment
thereof through
hapten, so as to be targeted to cells. The drug delivered to the target cell
may be a drug for
preventing, improving or treating PDGF or PDGF receptor-related diseases, for
example,
neovacular disorders.
An embodiment of the present invention is a pharmaceutical composition for
preventing, improving or treating neovascular disease, comprising an antibody-
drug conjugate
in which a drug is conjugated to the anti-PDGF receptor antibody or antigen-
binding fragment
22
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
thereof, or an antibody-drug conjugate in which a drug is conjugated to the
anti -PDGF receptor
antibody or antigen-binding fragment thereof through hapten.
An embodiment of the present invention relates to a method of treating a
subject
diagnosed as suffering from a neovacular disease or having a risk of
occurrence of a
neovacular disease , comprising a step of administering an antibody-drug
conjugate in which a
drug is conjugated to the anti-PDGF receptor antibody or antigen-binding
fragment thereof, or
an antibody-drug conjugate in which a drug is conjugated via hapten to the
anti-PDGF receptor
antibody or antigen-binding fragment thereof, to the subject at a sufficient
amount of
preventing, improving or treating the diseases in the subject as a first
therapeutic agent or an
auxiliary therapeutic agent.
In another embodiment, the pharmaceutical composition of the present invention

provides a means for preventing, improving, inhibiting or treating an ocular
neovascular
disease.
Ocular neovascular diseases that can be treated or suppressed by the
pharmaceutical
composition of the present invention include ischemic retinopathy, iris
neovascularization,
intraocular neovascularization, senile Age-related macular degeneration,
corneal
neovascularization, retinal neovascularization, choroidal neovascularization,
diabetic retinal
ischemia, or proliferative diabetic retinopathy.
Specifically, an animal model with the oxygen-induced retinal disease is an
animal
model of retinopathy of premature baby (A. Hellstrom, LE Smith, D. Dammann,
Retinopathy
of prematurity, Lancet. 382 (2013) 1445-57). An animal model with laser-
induced choroidal
neovascularization is an animal model of wet type senile macular degeneration
(V. Lambert, J.
Lecomte, S. Hansen, S. Blacher, ML. Gonzalez, et al, Laser-induced choroidal
23
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
neovascularization model to study age-related macular degeneration in mice,
Nat Protoc. 8
(2013) 2197-211). Both of them are diseases caused by an important mechanism
of
angiogenesis. Like proliferative diabetic retinopathy, it can also be used as
a therapeutic agent
for diseases such as proliferative diabetic retinopathy, in which angiogenesis
is an important
mechanism (Y. Qazi, Mediators of ocular angiogenesis, J. Genet. 88 (2009) 495-
515) ).
In another embodiment, the pharmaceutical composition of the present invention

provides a means for preventing, ameliorating, inhibiting or treating
neovascular diseases
neovascular disease such as cancer or tumor. The cancer is a cancer having a
problem with
PDGFR-f3, and includes retinoblastoma, glioma, testicular cancer, breast
cancer, ovarian cancer,
melanoma, lung cancer, and prostate cancer, but is not limited to.
In addition, the diseases having a problem with PDGFR-13 which shows
cytotoxicity
in NIH3T3 expressing mPDGFRb and human pericyte cells expressing hPDGFR-13
include
retinoblastoma (ZK Goldsmith et al., Invest.Ophthalmol. Vis.Sci. 59 (2018)
4486-4495),
glioma (I. Nazarenko et al., J. Med. Sci. 117 (2012) 99-112), colorectal
cancer (S. Fujino et al.,
Oncol. Rep. 39 (2018) 2178-2184), testicular cancer (S. Basciani et al.,
Endocrinology. 149
(2008) 6226-35), breast cancer (J. Paulsson et al., J. Pathol) Clin.Res. 3
(2017) 38-43), ovarian
cancer (S. Avril et al., Oncotarget. 8 (2017) 97851-97861), melanoma, lung
cancer, prostate
cancer (M. Raica et al., Pharmaceuticals (Base1s). 3 (2010) 572-599), and the
like.
As used herein, the terms -angiogenesis" and -neovascularization" are used
interchangeably. Angiogenesis and neovascularization refer to the development
of new blood
vessels into cells, tissues, or organs. The regulation of angiogenesis
typically changes in certain
disease states, and in many cases, the pathological damage associated with
this disease is
associated with altered, unregulated or uncontrolled angiogenesis. Persistent,
unregulated
24
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
angiogenesis occurs in a number of disease states including those
characterized by abnormal
growth of endothelial cells, and supports the pathological damage observed in
these conditions
including leakage and permeability of blood vessels.
In the present specification, -ocular neovascular disease" or -ocular
neovascular
disease" refers to a disease characterized by neovascularizsation that is not
changed or
controlled in the eye of a subject. Exemplary ocular neovascular diseases
include ischemic
retinopathy, iris angiogenesis, intraocular angiogenesis, senile macular
degeneration, corneal
angiogenesis, retinal angiogenesis, choroidal angiogenesis, diabetic retinal
ischemia,
proliferative diabetic retinopathy, and the like.
As used herein, -PDGF" or -platelet-derived growth factor" refers to a
mammalian
platelet-derived growth factor that affects neovascularization or angiogenesis
process. As used
herein, the term -PDGF" generally refers to a class of growth factors that
induce DNA synthesis
and mitosis through binding and activation of platelet-derived growth factor
cell surface
receptors (ie, PDGF receptors) to reactive cell types.
In the present invention, it is observed that the single-chain variable
fragment (scFv)
of an antibody against platelet-derived growth factor receptor beta (mPDGFR-
13)-kappa
constant region (Cx)-scFy fusion protein, and cotinine-duocarmycin form an
antibody-drug
conjugate complex to induce cytotoxicity against PDGFR-P-expressing cells. An
improved
approach for antibody selection in antibody-drug conjugate preparation is
demonstrated by
preparing a number of anti-mPDGFR-13 antibody candidates in the bispecific
scFv-Cx-scFy
fusion protein format and testing their ability to deliver cytotoxic drugs
binding to cotinine.
An embodiment of the present invention relates to a method of selecting an
antibody
that efficiently delivers a drug into a target cell using a hapten-drug
conjugate and a bispecific
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
antibody.
A further embodiment of the present invention relates to a drug-antibody
conjugate
comprising the selected antibody and a drug, or a complex comprising the
selected antibody
and a drug-hapten.
The hapten-drug conjugate used in the method of the present invention refers
to a
conjugate in which a target drug to be delivered to a target cell and a hapten
are connected. The
hapten is a substance that does not exist naturally in body to prevent binding
to cells, does not
de novo biosynthesized in body, does not cause physiological activity, and has
a chemical
functional group for effective binding with a substance to be conjugated with.
Hapten usable
in the present invention is not particularly limited, but for example, can be
selected from
cotinine, organic molecules with small molecular weight such as DNP (2,4-
dinitrophenol),
TNP (2,4,6-trinitrophenol), biotin, and digoxigenin.
As an example of the hapten, cotinine is an ideal hapten to be used in this
platform
because of exogeneity, non-toxicity, and physiological inei _________ iness as
a major metabolite of
nicotine. Trans-4-cotininecarboxylic acid can bind to several substances. The
cotinine-drug
conjugate can be easily synthesized, and has higher purity than that of the
existing antibody-
drug conjugate, and can be synthesized as a cotinine-drug conjugate having
various drugs and
linkers. In addition, since the drug is not directly conjugated to the
antibody, it does not affect
the affinity and stability of the antibody. The antibody-drug conjugates are
made by simply
reacting a bispecific antibody with a cotinine-drug conjugate.
Drugs applicable to the present invention are substances to be delivered to a
target
using antibodies and are not particularly limited, but for example, include
DNA synthesis
inhibitors (e.g., calicheamicin, pyrrolobenzodiazepine (PBD), duocarmyin
derivative,
26
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
anthracyclineNemorubicin, doxorubicin, Irinotecan, amatoxin), Microtubule
inhibitors (e.g.,
auristatin, maytansine, tubulysin), Topo-isomerase inhibitor (SN-38),
photosensitizer (e.g., 5-
Aminolaevulinic acid (ALA), Benzoporphyrin derivative monoacid ring A (BPD-
MA),
Chlorins, Tetra (m-hydroxyphenyl)chlorin (mTHPC), Lutetium texaphyrin), and
the like. The
photosensitizing agent alone does not exert its efficacy, but it is delivered
locally or
systemically to target cells or tissues or delivered, and activates the
delivered drug by
irradiating a laser to a specific area, for example, a lesion site such as
eyeball or eye, thereby
exerting the efficacy of the drug. Accordingly, the photosensitizing agent may
be conjugated
to an antibody and administered to a subject in need of, and then a step of
exerting a medicinal
effect by irradiating a laser may be additionally performed.
The method according to the present invention may use a bispecific antibody
that binds
specifically binds to the target antigen present in the target tissue or cell
to which the drug is
delivered, and can also specifically bind to the hapten included in the hapten-
drug conjugate.
For example, bispecific antibody applicable to the present invention may have
a structure of
first antibody-linker-second antibody, in which the first antibody and the
second antibody are
antibodies that specifically bind to a hapten or a target antigen,
respectively. The bispecific
antibody having the structure of the first antibody-linker-second antibody may
be a fusion
protein of the first antibody (scFv)-C¶kappa constant)-second antibody (scFv).
For example,
it can be connected in the order of VH-GQSSRSSGGGGSSGGGGS-VL from the N-
terminus,
and the linker connects C-terminus of VH to N-terminus of VL. It can be a
structure of (ScFv
of anti-PDGFR-13 antibody)-(GGGGS)3-Ck-(GGGGS)3-(scFv of anti-cotinine
antibody). An
amino acid sequence or nucleotide sequence of cotinine applicable to the
present invention is
widely known in the art, and for example, is referred to US8008448B.
27
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
In an embodiment of the present invention, when the target antigen is platelet-
derived
growth factor receptor beta (PDGFR-13) and the hapten is cotinine, the
bispecific antibody can
be an antibody in which an antibody against a target antigen (anti-PDGFR-13
antibody) and an
antibody against hapten (anti-hapten antibody) is linked with a linker, and
specifically, a fusion
protein having the structure of the first antibody (scFy of anti-PDGFR-13
antibody)-CK-the
second antibody (scFy of anti-hapten antibody).
The present inventors tested the feasibility of this platform using a
bispecific antibody
fusion protein of (scFy of anti-human platelet-derived growth factor receptor
beta (hPDGFRP)
x CI< x (scFy of anti-cotinine antibody). The present inventors constructed
monovalent or
bivalent cotinine-duocarmycin, which are named as cotinine-duocarmycin (cot-
duo) or
cotinine-duocarmycin-cotinine (cot-duo-cot) (Fig. lb). The present invention
indicates that the
platform can be used to screen optimal combination of antibodies and drug for
development of
antibody-drug conjugate, compared to conventional platform.
A complex including bispecific antibody (for example, anti-PDGFRP x cotinine
scFv-
CirscFv fusion protein) and hapten-drug conjugate (for example, cotinine-
conjugated
duocarmycin), shows specific cytotoxicity in NIH3T3 cell lines expressing
target antigen (for
example, mPDGFR-13). The present invention can be used to select the
combination of
internalizing antibody and drug in the development of antibody-drug conjugate.
For example,
four antibody clones bound to mPDGFR-13 coated in bottom in a dose-dependent
manner (Fig.
3a), while they had no reactivity against negative control-human Fc protein.
The present
inventors confirmed the binding activity of individual clones to NIH3T3 cell
lines expressing
28
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
mPDGFRP and found that biotin--mPDGF-BB bound to the cells with using a flow
cytometry
(Fig. 4b).
Antibody clones (PRb-CC01, PRb-CCO2, and PRb-CC03) against three types of
mouse antigens according to the present invention may bind to domain 2 or 3 of
mPDGFR-13
by competing with mPDGF-BB which is a ligand of mPDGFR-f3. One antibody clone
(PRb-
CNO1) may bind to domains 1, 4, or 5 instead of domains 2 and 3, because it
does not compete
with mPDGF-BB. The binding abilities of PRb-CC01, PRb-CCO2, and PRb-CCO3 to
mPDGFR-13 were inhibited by mPDGF-BB (Fig. 4a), but the binding ability of PRb-
CNO1 was
not inhibited by mPDGF-BB. The present inventors measured the cell binding
ability of each
clone using flow cytometry, when mPDGF-BB was present in NIH3T3 cells
expressing
mPDGFR-13. The present inventors found that the bispecific antibody of scFv-Cx-
scFy fusion
protein and biotin-mPDGF-BB bound to the cells. Like the result of competition
enzyme
immunoassay, only PRb-CNO1 was able to bind to mPDGFR-13 in the presence of
mPDGF-
BB-biotin (Fig. 4b), and the binding capacity of the other three clones was
blocked by mPDGF-
BB.
When the antibodies of the present invention were expressed in the form of a
bispecific
antibody of scFv-Cx-scFv, the bispecific antibodies were capable of binding to
both mPDGFR-
(3 and cotinine. In addition, the bispecific antibodies could be conjugated
with several cotinine
drugs such as cotinine-duocarmycin (cot-duo) and cotinine-duocarmycin-cotinine
(cot-duo-
cot). When conducting toxicity tests with complexes such as cot-duo and cot-
duo-cot, the
complexes are toxic. The four bispecific antibodies of scFv-Cx-scFy fusion
proteins (PRb-
CC01, PRb-CNO1, PRb-CCO2, PRb-CC03) were capable of simultaneously binding to
mPDGFR- (3 and cotinine, unlike the control bispecific antibody (FIG. 3d).
29
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
To evaluate the toxicity of the four types of bispecific antibodies of scFv-Cx-
scFv and
cotinine-duocarmycin conjugates, NIH3T3 cells were cultured together with the
conjugates
under two conditions with or without mPDGF-BB, and were tested for the
relative cell viability
using intracellular adenosine triphosphate (ATP). The toxicity of the PRb-CNO1
and cot-duo
complex or the PRb-CNO1 and cot-duo-cot complex was also high with the
addition of
mPDGF-BB, but toxicity rate of the fusion proteins (PRb-CC01, PRb-CCO2, PRb-
CC03)
competing with mPDGF-BB and cot-duo complex or cot-duo-cot complex decreased
with the
addition of mPDGF-BB (Fig. 5).
In the present invention, the scFv-Cx-scFv bispecific antibody fusion protein
and
cotinine-duocarmycin (cot-duo) were prepared respectively (Fig. lb), and then
conjugate of the
bispecific antibodies scFv-Cx-scFv and cotinine-duocarmycin was prepared and
performed for
the toxicity test. As a result, they showed characteristic toxicity in human
pericyte cells
expressing hPDGFR-fl. The result of the present invention indicates that it is
possible to select
an optimal antibody and drug combination for the development of antibody-drug
conjugates
compared to the convention platforms.
For example, it was shown that three antibody clones binding to hPDGFR-fl of
an
example of a target antigen according to the present invention, bound to
hPDGFR-fl in a
concentration-dependent manner. The three fusion proteins were concentration-
dependently
bound to hPDGFR-fl coated on the bottom (Fig. 12a), while the negative control
did not bind
to the coated human Fc protein (Fig. 12b). In the present invention, it was
confirmed that the
ability of each clone binding to human pericyte cells expressing hPDGFR-fl in
the presence of
hPDGF-BB was analyzed using a flow cytometer, resulting in biotin-hPDGF-BB
bound to the
cells (Fig. 13b).
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
The three types of antibody clones against hPDGFR-13 according to the present
invention (PRb-CN16, PRb-CN32, PRb-CN26) did not compete with hPDGF-BB of a
ligand
of hPDGFR-13, and thus was expected to bind to domains 1, 4 or 5 instead of
domains 2 and 3.
The binding ability of PRb-CN16, PRb-CN32, and PRb-CN26 to hPDGFR-13 was not
inhibited
by hPDGF-BB (Fig. 13a).
In the present invention, when hPDGF-BB is present in human pericyte cells
expressing hPDGFR-13, the cell binding ability of each clone was measured
using flow
cytometry. It was confirmed that the bispecific antibody of scFv-Cx-scFv
fusion protein and
biotin-hPDGF-BB were bound. Like the result of competitive enzyme immunoassay
method,
three kinds of antibody clones (PRb-CN16, PRb-CN32, PRb-CN26) were able to
bind to
hPDGFR-13 in the presence of hPDGF-BB-biotin (Fig. 13b). When the antibody
against the
target antigen according to the present invention is expressed in the form of
a bispecific
antibody of scFv-Cx-scFv, the bispecific antibody can bind to both the target
antigen and the
hapten. In addition, the bispecific antibodies can be complexed with several
cotinine-drug
conjugates such as cotinine-duocarmycin (cot-duo). When conducting toxicity
tests with the
conjugates such as cot-duo, the complexes are toxic.
The three bispecific antibodies of scFv-Cx-scFv fusion proteins (PRb-CN16, PRb-

CN32, PRb-CN26) against human PDGFR-13 according to the present invention were
capable
of simultaneously binding hPDGFR-13 and cotinine unlike the control bispecific
antibody (Fig.
12d). To evaluate the toxicity of the bispecific antibody of scFv-Cx-scFv and
cotinine-
duocamycin conjugates prepared by using three types of hPDGFR-13 antibodies,
human
pericyte cells were cultured with the conjugates under two different
conditions with or without
hPDGF-BB, and the relative cell viability was measured with intracellular
adenosine
31
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
triphosphate (ATP). The toxicity of the complexes consisting of anti-hPDGFR-
Pantibody of
PRb-CN16, PRb-CN32, PRb-CN26, and the cot-duo was also high with the addition
of
hPDGF-BB (FIGS. 14a and 14b).
The present inventors tested a new platform for developing antibody-drug
conjugate
consisting of bispecific scFv-C-K-scFv fusion protein and cotinine-cytotoxic
drug, which
simplifies the process for selecting antibody and drug for the optimal
antibody-drug conjugate.
Ideally, it is practical to select antibodies that ensure efficient
internalization and cytotoxic drug
release. However, since the production of individual specific antibody-drug
conjugate
molecules using candidate antibodies requires considerable work to optimize
and characterize
including DAR (drug-antibody ratio), antibodies have been screened based on
the speed and
efficiency of internalization in the initial screening step. Moreover, as the
process by which
antibody-drug conjugates act on cells is understood better, it can be seen
that internalization of
antibodies is not the only factor determining the efficacy of antibody-drug
conjugates.
Cotinine is not toxic and has an LD50 value of 4 0.1 g/kg in rats. In
addition, no
adverse side effects were induced in persons treated with up to 1,800 mg of
cotinine for 4
consecutive days. The present inventors have observed that there is a slight
difference between
the cytotoxicity of free duocarmycin and cotinine-duocarmycin conjugates in an
ex vivo
environment. The formation of a complex between the anti-HER2 antibody x
cotinine of scFv-
C-K-scFv fusion protein and the cotinine-duocarmycin conjugate lowers the
toxicity of
duocaramycin in the ex vivo environment. It implies that the bispecific
antibody forms a
complex with the drug and decreases the absorption of the drug into the cell.
In conventional
studies, it was reported that duocarmycin induced significantly weight loss in
mice. However
in vivo experiment using a tetravalent bispecific antibody of the present
invention, there was
32
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
no significant weight loss in mice which a cotinine-duocarmycin conjugate
mixed with a
negative control IgG was injected into. This fact opens up the possibility of
introducing various
types of linkers between cotinine and a toxic drug (duocarmycin), and
releasing toxins from
cotinine only in the tumor environment like metalloproteases. Because the
cotinine-drug
conjugate is released without degradation in the tumor environment, it will be
quickly removed
from the blood flow without damaging normal cells.
Even after the addition of mPDGF-BB, the cytotoxicity of PRb-CNO1-containing
bispecific antibody complexed with cot-duo or cot-duo-cot was still high, but
the addition of
mPDGF-BB, which promotes cell proliferation, the toxicity of the three
antibodies (PRb-CC01,
PRb-CCO2 and PRb-CC03), which competes with mPDGF-BB, complexed with either
cot-duo
or cot-duo-cot was slightly reduced. mPDGF-BB may strengthen the
internalization of receptor
by binding to PDGFR-fl which has been already bound to PRb-CN01, thereby
promoting the
cytotoxic effects of cot-duo and cot-duo-cot. In the present invention, the
PRb-CNO1 construct
did not compete with PDGF-BB and induced cytotoxicity independently of PDGF-BB
(FIGS.
5a to 5d). PDGFR-fl is highly associated with angiogenesis, and is mainly
overexpressed in
pericytes.
[Effect of the Invention]
In the antibody against the PDGF receptor according to the present invention,
the
antibody-drug conjugate in which a chemotherapeutic agent is conjugated to the
antibody and
the use of preventing or treating ocular neovascular diseases by using the
same, the antibody
against the PDGF receptor exhibits more toxicity specifically at a certain
concentration to
pathological cells or tissues that express a lot of hPDGFR-fl, while
minimizing toxicity to
normal cells or normal tissues, thereby being applicable for an excellent
therapeutic agent. In
33
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
addition, the antibody against the PDGF receptor has an advantage that the
internalization level
of the antibody is not suppressed or is increased, even if the expression
level of PDGFR-13
and/or PDGF-BB increases.
[Brief description of drawings]
Fig. la and Fig. lb are diagrams showing a bispecific antibody related to an
anti-
mPDGFR-13 antibody and cotinine-duocarmycin according to an example of the
present
invention.
Fig. 2 shows the result of SDS-polyacrylamide gel electrophoresis of the
bispecific
antibody related with the anti-mPDGFR-13 antibody according to an example of
the present
invention. Lane 1, reduced PRb-CCO3 x cotinine; Lane 2, non-reduced PRb-CCO3 x
cotinine;
Lane 3, reduced PRb-CCO1 x cotinine; Lane 4, non-reduced PRb-CCO1 x cotinine;
Lane 5,
reduced PRb-CNO1 x cotinine; Lane 6, non-reduced PRb-CNO1 x cotinine; Lane 7,
reduced
PRb-CCO2 x cotinine; Lane 8, non-reduced PRb-CCO2 x cotinine; Lane 9, reduced
anti-HER2
x cotinine (control); Lane 10, non-reduced anti-HER2 x cotinine (control). M:
molecular
weight marker.
Fig. 3a is a graph showing the binding ability of the bispecific antibody
related with
the anti-mPDGFR-13 antibody according to an embodiment of the present
invention, in which
the bispecific antibody PRb-CCO1 (0), PRb-CNO1 (.),PRb-CCO2 (1), PRb-CCO3 (T)
and a
negative control (*) were incubated on a plate coated with mPDGFR-13 chimera.
Each well
was reacted with IIRP-anti human CI< antibody and TMB. The results show the
mean value
standard error of the duplicate experiments. Fig. 3b shows the TNF-cc receptor
extracellular
region-human Fc fusion protein used as a negative control antigen for enzyme
immunoassay.
In Fig. 3c, in order to confirm the binding ability of the bispecific antibody
scFv-Cx-scFy
.5 4
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
fusion protein to cotinine, the bispecific antibody scFv-Cx-scFy was reacted
to the plate coated
with cotinine-BSA, and reacted with HRP-anti-human CI< antibody and TMB. In
Fig. 3d, in
order to confirm the simultaneous binding ability of the bispecific antibody
scFv-Cx-scFy
fusion protein to cotinine and mPDGFR-13, mPDGFR-13-Fc chimera was reacted to
the plate
coated with cotinine-BSA, and HRP-anti-human Fc Antibodies and TMB were
treated. The
results show the mean value standard error of three repetitive experiments.
***p < 0.001 was
compared to the control group.
Fig. 4a to Fig. 4c are graphs confirming competition and intracellular
internalization
of PDGF-BB and bispecific antibodies related with anti-mPDGFR-13 antibody. In
Fig. 4a, the
PDGFR-P-Fc chimera coated plate was reacted with scFv-Cx-scFy in the absence
(left) or
presence (right) of mPDGF-BB (100 nM) and then the bound bispecific antibody
fusion Protein
was measured by HRP-anti-human CI< antibody and TMB. In Fig. 4b, the
bispecific antibody
scFv-Cx-scFy fusion protein (100 nM) in the flow cytometry buffer was reacted
to NIH3T3
cells expressing mPDGFR-13 with or without mPDGF-BB-biotin. The cells were
treated with
APC-anti human CI< antibody and streptavidin-PE. The bispecific antibody anti-
HER2 x
cotinine scFv-Cx-scFy fusion protein was used as a negative control. The
internalization of the
bispecific antibody scFv-Cx-scFy fusion protein was visualized with confocal
microscope.
After NIH3T3 cells were treated by the fusion protein, antibodies bound on the
cell surface
were removed. After cell fixation, the fusion protein was stained with FITC -
anti human CI<
antibody (green). To image early endosomes, the cells were stained with anti-
Rab5 antibody
and Alexa Fluor 546-goat anti-rabbit antibody IgG (red). The part indicated by
the arrow shows
the enlarged part in which the fluorescence of the anti-PDGFR-13 x cotinine
scFv-Cx-scFy
fusion protein and the initial endosome are co-localized. DNA was stained with
DAPI (blue)
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
(Scale bar, 10 pm)
Fig. 5a to Fig. 5d are graphs confirming the cytotoxic ability of the
bispecific antibody
against anti-mPDGFR-13 and cotinine-duocarmycin complexes to cells expressing
PDGFR-P.
Fig. 5a shows that NIH3T3 cells were treated with a bispecific antibody scFv-
Cx-scFv fusion
protein and a cot-duo complex without mPDGF-BB. Cellular ATP was measured to
assess
relative cell viability. The bispecific antibody anti-HER2 x cotinine scFv-Cx-
scFv fusion
protein was used as a negative control. In Fig. 5b, the experiment was
repeated in the presence
of mPDGF-BB. In Fig. 5c, NIH3T3 cells were treated with a complex of
bispecific antibody
and cot-duo-cot without mPDGF-BB. In Fig. 5d, the experiment was repeated in
the presence
of mPDGF-BB. DMSO was used as a vehicle control for cot-duo and cot-duo-cot.
The results
show the mean value standard error of three repetitive experiments.
Fig. 6 is a graph showing the experimental result of the binding ability of a
bispecific
antibody related with an anti-mPDGFR-13 antibody according to the present
invention to
MOLT-4 cells not expressing PDGFR-13 with a flow cytometer. MOLT-4 cells were
treated
with the bispecific antibody (100 nM) and stained with an APC-anti human CI<
antibody (clone
TB28-2, BD Biosciences, San Jose, CA, USA).
Fig. 7 is a graph confirming the cytotoxic ability of the bispecific antibody
related to
the anti-mPDGFR-13 antibody and cot-duo complexes in the cells not expressing
PDGFR-P. In
Fig. 7a, MOLT-4 cells were reacted with the bispecific antibody and cot-duo
complex (DAR4).
The cellular ATP was measured to evaluate the relative cell viability. In Fig.
7b, MOLT-4 cells
were reacted with the bispecific antibody and cot-duo complex (DAR2). The
bispecific
antibody anti-HER2 x cotinine scFv-Cx-scFv was used as a negative control.
DMSO was used
as a vehicle control for cot-duo. The results show the mean value standard
error of three
36
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
repetitive experiments.
Fig. 8 shows a result of size exclusion chromatography of a bispecific
antibody
containing an anti-mPDGFR-13 antibody according to the present invention. The
bispecific
antibodies, the complex of bispecific antibodies and cot-duo, the complex of
bispecific
antibodies and cot-duo-cot according to the present invention were analyzed
with the size
exclusion chromatography-HPLC using Dionex Ultimate 3000 equipped with a Sepax
SRT-C
SEC-300 column. The mobile phase was used as PBS, and the mobile solvent was
eluted at 1
mL/min for 15 minutes. The ultraviolet detector was adjusted to 254 nm and the
results were
monitored by mAU. HMW is a high molecular weight species.
Fig. 9 is a result showing that the bispecific antibody related to the anti-
mPDGFR-13
antibody and cot-duo complexes according to the present invention inhibit
angiogenesis in an
oxygen-induced retinal disease animal model.
Fig. 10 shows that the bispecific antibody related with anti-mPDGFR-13
antibody and
cot-duo complexes according to the present invention inhibits angiogenesis in
a laser-induced
choroidal neovascularization animal model.
Fig. 11 is a result of SDS-polyacrylamide gel electrophoresis of a cotinine
scFv-Cx-
scFv fusion protein as a bispecific antibody related with the anti-hPDGFR-13
antibody
according to the present invention: Lane 1, reduced PRb-CN16 x cotinine; Lane
2, non-reduced
PRb-CN16 x cotinine; Lane 3, reduced PRb-CN26 x cotinine; Lane 4, non-reduced
PRb-CN26
x cotinine; Lane 5, reduced PRb-CN32 x cotinine; Lane 6, non-reduced PRb-CN32
x cotinine;
Lane 7, reduced anti-mCD154 x cotinine (control); Lane 8, non-reduced anti-
mCD154 x
cotinine (control); M, molecular weight marker.
In Fig. 12a, the bispecific antibodies of scFv-Cx-scFv fusion protein such as
PRb-
37
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
CN16 (0) PRb-CN32 (N), and PRb-CN26 (1) and the negative control (*), and
cotinine scFv-
Ck-ScFv fusion protein were reacted at various concentrations on hPDGFRP
chimera-coated
microtiter plates. Fig. 12b is a result of reaction of HRP-anti-human CI<
antibody and TMB in
each well, in which the results are shown as mean value standard error from
the duplicate
experiments. In Fig. 12c, TNF-a receptor extracellular domain-human Fc fusion
protein was
used as a negative control antigen in an enzyme immunoassay. In Fig. 12d, in
order to confirm
the binding ability of the bispecific antibody related with the anti-hPDGFR-13
antibody
according to the present invention, the bispecific antibody scFv-C-k-scFv
fusion protein was
cultured on a plate coated with cotinine-BSA, and was treated with HRP-
conjugated anti-
human Ci, antibody and TMB. In order to confirm the simultaneous binding of
bispecific scFv-
CirscFv fusion proteins to cotinine and hPDGFRP, hPDGFRP-Fc chimera was
cultured on
microtiter wells coated with cotinine-BSA and was treated with HRP-conjugated
anti-human
Fc antibody, and TMB. The results are shown as the mean SD from triplicate
experiments.
***p <0.001 is compared to controls.
Fig. 13a to Fig. 13d are graphs confirming the competition and the cellular
internalization of the bispecific antibody related with anti-hPDGFRP according
to the present
invention with PDGF-BB. In Fig 13a, bispecific scFv-CirscFv fusion proteins
were incubated
on the hPDGFRP-Fc chimera-coated microtiter plates without (left) or with
(right) hPDGF-BB
(100 nM), and the amount of bound bispecific antibody fusion protein was
determined using
HRP-conjugated anti-human Ci, antibody and TMB. In Fig. 13b, human pericyte
cells
expressing hPDGFR-13 were treated with bispecific scFv-CirscFv fusion proteins
(100 nM) in
flow cytometric assay buffer without or with hPDGF-BB-biotin. The cells were
probed with
APC-conjugated anti-human Ci, antibody and streptavidin-PE. Bispecific anti-
mCD154 x
38
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
cotinine scFv-CirscFy fusion protein was used as a negative control. In Fig.
13c, the confocal
microscopy visualized the internalization of bispecific scFv-CirscFy fusion
protein. After
human pericyte cells were incubated with fusion proteins, surface-bound
antibodies were
removed. After cell fixation, the fusion proteins were stained with FITC -
conjugated anti-
human Ci, antibody (green). To image the early endosome, the cells were
incubated with anti-
Rab5 antibodies and Alexa Fluor 546-conjugated goat anti-rabbit IgG (red). The
part indicated
shows the enlarged part which shows co-localization of anti-hPDGFR-13 x
cotinine scFv-Cir
scFy fusion proteins and early endosomes. DNA was stained with DAPI (blue).
Images were
merged after initial capture. Scale bar, 10 p.m. Fig. 3d shows that assay was
repeated with the
addition of hPDGF-BB.
Fig. 14a and Fig. 14b are graphs showing that cytotoxicity assays of
bispecific
antibody related with anti-hPDGFRP antibody according to the present invention
complexed
with cot-duo on PDGFRP-expressing cells. In Fig. 14a, human pericyte cells
were treated with
the bispecific scFv-Cx-scFy fusion protein and cot-duo complex. In Fig. 14b,
the assay was
repeated with the addition of hPDGF-BB. The cellular ATP levels were measured
to determine
relative cell viability. The bispecific anti-mCD154 x cotinine scFv-Cx-scFy
fusion protein was
used as a negative control. DMSO was used as a vehicle control for cot-duo.
Results are shown
as the mean SD from three repetitive experiments.
Fig. 15 is a graph showing Flow cytometry analysis on reactivity of bispecific
antibody
reacted with anti-hPDGFR-13 antibody on the cells not expressing hPDGFR-13. A-
431 cells were
incubated with bispecific anti-hPDGFR-13 x cotinine scFy-Cx-scFy fusion
proteins (100 nM)
and probed with APC-conjugated anti-human CI< antibody (clone TB28-2, BD
Biosciences,
San Jose, CA, USA).
39
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
Fig. 16 is a graph showing that cytotoxicity assays of bispecific antibody
related with
anti-hPDGFR-13 antibody complexed with cot-duo complexes on the cells not
expressing
hPDGFR-f3. A-431 cells were treated with anti-hPDGFR-13 x cotinine fusion
protein and cot-
duo (DAR4). The cellular ATP levels were measured to determine relative cell
viability.
Bispecific anti-mCD154 x cotinine scFv-Cic-scFy fusion protein was used as a
negative control.
DMSO was used as a vehicle control of cot-duo. Results are shown as the mean
SD acquired
from three repetitive experiments.
Fig. 17 is a graph showing cross-species test of bispecific antibody related
with anti-
hPDGFR-13 antibody. hPDGFR-13, mPDGFR-13, Rhesus PDGFRP, Cynomolgus monkey
PDGFRP. Sus scrofa PDGFRP and control Fc coated microtiter plates were
incubated with
bispecific antibody containing anti-hPDGFRP antibody (100 nM) and the amount
of bound
bispecific fusion protein was determined using HRP-conjugated anti-human Ci,
antibody and
TMB.
Fig. 18 shows that the bispecific antibody scFv-Cic-scFy fusion protein (100
nM) in
flow cytometry buffer was incubated on NIH3T3 cells expressing mPDGFR-f3. The
cells were
treated with APC -anti human CI< antibody. The bispecific antibody anti-HER2 x
cotinine scFv-
Cic-scFv fusion protein was used as a negative control.
Fig. 19a to Fig. 19b are results confirming the competition for PRb-CN01
surrogate
antibody. In Fig. 19a, various concentrations of PRb-CN01-rabbit Fc (0.01 nM-1
04), PRb-
CN01, anti-HER2 control antibody, and PRb-CN32 bispecific antibody scFv-Cic-
scFy (100 nM)
diluted in 3% BSA/PBS were reacted on the plate coated with mPDGFR-13-hFc
chimeric
protein., and were reacted at each well at 37 C for 2 hours. Then, the bound
PRb-CNO1-rabbit
Fc protein was measured with HRP-anti rabbit Fc antibody and ABTS. In Fig.
19b, the
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
mPDGFR-13-expressing NIH3T3 cells were reacted with PRb-CNO1-rabbit Fc (50 nM)
and
PRb-CN01, a control antibody, and PRb-CN32 bispecific antibody scFv-Cx-scFv (1
pM). The
cells were incubated with FITC-anti rabbit Fc antibody. The bispecific
antibody anti-HER2 x
cotinine scFv-Cx-scFv fusion protein was used as a negative control.
Fig. 20a to Fig. 20b are graphs confirming the difference in cytotoxic
activity of PRb-
CNO1 and PRb-CN32 bispecific antibodies and cotinine-duocarmycin complexes in
the cells
expressing PDGFR-f3. In Fig. 20a, NIH3T3 cells were treated with a bispecific
antibody scFv-
Cx-scFv fusion protein and a cot-duo complex without mPDGF-BB. The cellular
ATP was
measured to assess relative cell viability. The bispecific antibody anti-HER2
x cotinine scFv-
Cx-scFv fusion protein was used as a negative control. In Fig. 20b, the
experiment was repeated
in the presence of mPDGF-BB.
[Mode for Invention]
The present invention will be described in more detail with reference to the
following
examples, but the scope of the present invention is not intended to be limited
to the following
examples.
Example 1. Expression and purification of mPDGFR-I3 and Cu fusion proteins
The extracellular region of mPDGFR-13 is a peptide having an amino acid
sequence of
SEQ ID NO: 60 in the following table and a nucleotide sequence encoding it was
prepared, and
were chemically synthesized by adding a Sfi/ endonuclease recognition sites
(GenScript
Biotech, Jiangsu, China) to the end of the nucleotide. It was digested with
Sfi/ enzyme and
cloned into pCEP4 vector to prepare a recombinant pCEP4 expression vector,
which was
41
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
expressed by a previously reported method [Y. Lee, H. Kim et al., Exp. Mol.
Med. 46 (2014)
e114].
[Table 3]
SEQ ID
name An amino acid sequence
NO
MRLPGAMPALALKGELLLLSLLLLLEPQISQGLVVT 60
PPGPELVLNVSSTFVLTCSGSAPVVWERMSQEPPQE
MAKAQDGTFSSVLTLTNLTGLDTGEYFCTHNDSRG
LETDERKRLYIFVPDPTVGFLPNDAEELFIFLTEITEI
TIPCRVTDPQLVVTLHEKKGDVALPVPYDHQRGFS
GIFEDRSYICKTTIGDREVDSDAYYVYRLQVSSINV
SVNAVQTVVRQGENITLMCIVIGNEVVNFEWTYPR
hPDGFR-13 KESGRLVEPVTDFLLDMPYHIRSILHIPSAELEDSGT
YTCNVTESVNDHQDEKAINITVVESGYVRLLGEVG
TLQFAELHRSRTLQVVFEAYPPPTVLWFKDNRTLG
DSSAGEIALSTRNVSETRYVSELTLVRVKVAEAGH
YTMRAFHEDAEVQLSFQLQINVPVRVLELSESHPD
SGEQTVRCRGRGMPQPNIIWSACRDLKRCPRELPPT
LLGNSSEEESQLETNVTYWEEEQEFEVVSTLRLQH
VDRPLSVRCTLRNAVGQDTQEVIVVPHSLPFK
LVITPPGPEFVLNISSTFVLTCSGSAPVMWEQMSQV 61
PWQEAAMNQDGTFSSVLTLTNVTGGDTGEYFCVY
NNSLGPELSERKRIYIFVPDPTMGFLPMDSEDLFIFV
TDVTETTIPCRVTDPQLEVTLHEKKVDIPLHVPYDH
QRGFTGTFEDKTYICKTTIGDREVDSDTYYVYSLQ
VSSINVSVNAVQTVVRQGESITIRCIVMGNDVVNFQ
WTYPRMKSGRLVEPVTDYLFGVPSRIGSILHIPTAE
mPDGFR-f3 LSD SGTYTCNVSVSVNDHGDEKAINISVIENGYVRL
LETLGDVEIAELHRSRTLRVVFEAYPMPSVLWLKD
NRTLGDSGAGELVLSTRNMSETRYVSELILVRVKV
SEAGYYTMRAFHEDDEVQLSFKLQVNVPVRVLEL
SESHPANGEQTIRCRGRGMPQPNVTWSTCRDLKRC
PRKLSPTPLGNSSKEESQLETNVTFWEEDQEYEVVS
TLRLRHVDQPLSVRCMLQNSMGGDSQEVTVVPHS
LPFK
MRLPGAMPALALKGQLLLLPLLLLLEPQVSQGLVI 62
TPPGPELILNVSSTFVLTCSGSAPVVWERMSQELPQ
EMAKAQDNTFSSVLTLTNLTGLDTGEYFCTYNDSR
GLEPDERKRLYIFVPDPTVGFLPNDAEELFIFLTEITE
Cynomolgus ITIPCRVTDPQLVVTLHEKKGDIALPVPYDHQRGFS
Monkey GIFEDRSYICKTTIGDREVDSDAYYVYRLQVSSINV
PDGFR-f3 SVNAVQTVVRQGENITLMCIVIGNEVVNFEWMYP
RKESGRLVEPVTDFLLDMPYHIRSILHIPSAELEDSG
TYTCNVTESVNDHQDEKAINITVVESGYVRLLGEV
GALQFAELHRSRTLQVVFEAYPPPTVLWFKDNRTL
GDSSAGEIALSTRNVSETRYVSELTLVRVKVAEAG
42
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
HYTMRAFHEDAEVQ LS FQ LQ INVPVRVLEL SE SHP
DSGEQTVRCRGRGMPQPNIIWSACRDLKRCPRELP
PMLLGNSSEEESQ LETNVTYWEEEQEFEVVSTLRL
QHVDRPLSVRCTLRNAVGQDMQEVIVVPHSLPFK
MQLPRAVPASVLIGQVLLLPPLLLLGPQASWGLVIT 63
PP GPE LVLNL S STFVLTC S GPAPVVWERM SQKPPQE
MTGTQDGTFS SVLTLANVTGLDTGEYFCTYKGSPG
LEASERKRLYIFVPDPAVGFLPVDPEELFIFLTEITET
TIPCRVTDPRLVVTLHEKKVDVPLPISYDHQRGF SG
TFEDKTYVCKTTIGDREVDSDAYYVYSLQVS SINV
S f a SVGAVQTVVRQGENITVMCIVTGNEVVNFEWTYP
PDGFR us scro
RLETGRLVEPVTDFLFEMPHIRSI LHIP SAELGD S GT
-f3
YICNVSESVSDHRDEKAINVTVVESGYVRLIGELDA
VQFAELHRSRTLQVVFEAYPPPTVIWFKDNRTLGD
SGAGEIALSTRNVSETRYVSELTLVRVKVAEAGRY
TMRAFHEDAEAQISFQLQVNVPVRVLELSESHPAS
GEQTVRCRGRGMPQPHLTWSTC SD LKRCPRELPPT
PLGNSSEEESQLETNVTYWPQEQEFEVVSTLRLRR
VDQPLSVRCTLHNLLGHDAQEVTVVPHSLPFQ
Specifically, the recombinant pCEP4 expression vector was transduced into
human
fetal kidney 293F cell line (Invitrogen, Carlsbad, CA, USA) by using
polyethyleneimine
(Polysciences, Warrington, PA, USA) according to the previously reported
method [S.E. Reed
et al., J. Vriol. Methods. 138 (2006) 85-98]. The transduced cells were
cultured in GIBCO
Freestyle 293 expression medium containing 10,000 IU/L of penicillin and 100
mg/L of
streptomycin [S. Yoon et al., J. Cancer. Res. Clin. Oncol. 140 (2014) 227-33].
In six days after
transduction, the culture supernatant was collected and mPDGFR-13-CK fusion
protein was
purified by affinity chromatography using KappaSelect resin (GE Healthcare,
Buckinghamshire, UK) according to the manufacturer's instructions. In the
fusion protein, the
mPDGFR-13 amino acid sequence is from Leu at 32'd residue to Lys at 530th
residue of
mPDGFR-f3, which corresponds to the extracellular region, and is linked to CI<
at the C-
terminus of the mPDGFR-13 amino acid sequence through a linker-(GGGGS)3.
Example 2. Expression and purification of bispecific antibody (scFv-C.-scFv)
43
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
2-1. Preparation of combinatorial scFv-displayed phage library and bio-panning

Anti-mPDGFR-13 antibodies were selected from a phage library expressing scFvs
on
the surface made from immunized chickens.
Chickens were immunized at total 4 times with the mPDGFR-13-Cx fusion protein
obtained in Example 1 above. Serum obtained from blood collection from wing
veins before
and during immunization was used to test the animal's immune state by using
enzyme
immunoassay and flow cytometry. One week after the fourth immunization, they
were
sacrificed, and the spleen, bursa of Fabricius, and bone marrow were
harvested, and RNA was
isolated using a TRI reagent (Invitrogen).
Using the isolated RNA as a template, cDNA was synthesized using Superscript
III
First-Strand Synthesis system (Invitrogen), and then a phage library
expressing scFv was
produced according to the previously reported method [M.S. Lee et al.,
Hybridoma (Larchmt).
27 (2008) 18-241. That is, four phage libraries expressing 7.5 x 108, 6.9 x
108, 2.1 x 109, and
2.2 x 109 scFv were prepared using the cDNA. The biopanning was performed at
total five
times on the magnetic beads to which mPDGFR-13-Cx was bound with the library
[Y. Lee et
al., Exp. Mol. Med. 46 (2014) e 114].
At the 5th output titer, scFv clones were randomly selected and phage enzyme
immunoassay was performed on mPDGFR-P-Cx-coated microtiter plates (3690;
Corning Life
Sciences, Corning, NY, USA) [C.F. Barbas III, D.R. Burton, J.K. Scott, G.J.
Silverman, Phage
display- a laboratory manual, Cold Spring Harbor Laboratory Press, New York,
20011.
Clones with binding capacity to mPDGFR-13-Cx (A405> 1.5) were asked Macrogen
Inc. to perform sequence analysis with OmpSeq primers [W. Yang et al., Exp.
Mol. Med. 49
(2017) e3081. After analyzing an amino acid sequence, a total of 9 types of
antibody clones
44
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
were found (Table 3). In consideration of the expression rate and binding
ability, a total of four
clones (PRb-CN01, PRb-CC01, PRb-CCO2, PRb-CC03) were selected, and were
performed
for the subsequent experiments.
Table 4 below shows H-CDR3 amino acid sequence of anti-mPDGFR-13 scFv clones.
VH and VL and their CDR amino acid sequence information for the obtained four
clones are
shown in Table 1.
[Table 4]
Clone H-CDR3 sequences SEQ ID NO
PRb-CNO1 GVGSWAHGGRIDA 3
PRb-CCO1 GGGSIDA 11
PRb-CCO2 GYAGTIDA 19
PRb-CCO3 SSYIDT 27
2-2: Expression and purification of a bispecific antibody (scFv-Cu-scFv)
Using the anti-mPDGFR-13 scFv clone prepared in Example 1, pCEP4 expression
vector containing a gene encoding a (anti-mPDGFR-f3 scFv)-C-K-(anti-cotinine
scFv) bispecific
antibody was prepared.
The specific structure of the prepared (anti-mPDGFR-13 scFv)-C-K-(anti-
cotinine scFv)
bispecific antibody and the binding positions of each linker are shown in FIG.
la. Specifically,
the anti-mPDGFR43 scFv and anti-cotinine scFv are each joined in the order of
VL-
GQSSRSSGGGGSSGGGGS (SEQ ID NO: 57 in Table 2)-VH from the N terminal, and that

is, the C terminal of VL and the N terminal of VH are connected. From the N-
terminus, anti-
mPDGFR-13 scFv, Ck and anti -cotinine scFv are linked using each linker, and
the linker
connecting the scFv is (GGGGS)3 of SEQ ID NO: 59 in Table 2, and Ck amino
acids is shown
in SEQ ID NO: 58 in Table 2.
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
As a specific method of preparing a fusion protein of a bispecific antibody,
the gene
encoding anti-mPDGFR-13 scFv and the gene encoding anti-cotinine scfv were
digested with
Sill, Agel, and Notl (New England Biolabs) and ligated to the expression
vector. The gene
encoding the anti-mPDGFR-f3scFv was linked from the N-terminus to the C-
terminus in the
order of VL-linker-VH in Table 1, and the linker was a peptide prepared by
using the sequence
of SEQ ID NO: 57 in Table 2, to obtain a nucleotide sequence encoding it. The
gene encoding
the anti-cotinine scfv was obtained based on the contents described in
US8008448B, and the
obtained amino acid sequence of the cotinine is shown in the following table.
[Table 5]
Part Amino acid sequence SEQ ID
NO
CDR1-VH GHLRRRDWM 64
CDR2-VH IGRSGDT 65
CDR3-VH IPYFGWNNGD I 66
CDR1-VL QSSQ SPYSNEWLS 67
CDR2-VL RISTLAS 68
CDR3-VL AGGYNFGLFLFG 69
VH
EVQLVESGGGLVQPGGSLRLSCAASGHLRRR 70
DWMNWVRQAPGKGLEWVAAIGRSGDTYYA
TWAKGRFTISADTSKNTAYLQMNSLRAEDTA
VYYC SRIPYFGWNNGDIWGQGTLVTVSS
VL DIQMTQSPSSLSASVGDRVTITCQSSQSPYSNE 71
WLSWYQQKPGKAPKLLIYRISTLASGVPSRFS
GSRSGTDFTLTISSLQPEDFATYYCAGGYNFG
LFLFGQGTKVEIK
Trastuzumab scFv was cloned into an expression vector as a control for anti-
mPDGFR-
(3 scFv. Cysteine at the C-terminal portion of CI< was excluded to remove the
dimerization by
disulfide bonds. After transducing the obtained DNA construct into HEK293F
cells, a scFv-
C-K-scFv fusion protein was produced, and a bispecific antibody (scFv-C-K-
scFv) was purified
by affinity chromatography using KappaSelect resin.
46
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
To confirm the protein purity of the expressed bispecific antibodies, SDS-
polyacrylamide gel electrophoresis was performed using NuPage 4-12% Bis-Tris
gel
(Invitrogen) according to the manufacturer's instructions. 1 lig of protein
was added to the LDS
sample buffer with or without a reducing agent, and then reacted at 95 C for
10 minutes. Then,
electrophoresis was performed, and the gel was stained with Ezway Protein-Blue
II staining
solution (Koma Biotech, Seoul, Korea) to visualize the protein. SDS-
polyacrylamide gel
electrophoresis was performed (Fig. 2). A photograph of the result of the
electrophoresis is
shown in FIG. 2.
As shown in FIG. 2, the protein reduced by using a reducing agent was 67 kDa,
and
the non-reduced protein was stained at 60 kDa. The molecular weight of the
fusion protein
calculated by using the computer was 66.77 kDa. No multimeric bands were seen.
It was
expected that the non-reduced protein moved faster than the reduced protein,
because the non-
reduced protein was less resistant to the movement on the gel due to its dense
intrinsic
morphology.
In SEC-HPLC (FIG. 8), which is a result of size exclusion chromatography of a
bispecific antibody containing an antibody against mPDGFR-13 according to an
embodiment of
the present invention, PRb-CC01, PRb-CCO2, and PRb-CCO3 all showed monomeric
and
trimeric bands unlike PRb-CN01. When their conjugates were formed with cot-
duo, a large
amount of high molecular weight species (HMWs) were observed in all of PRb-
CC01, PRb-
CCO2, and PRb-CC03. In the case of PRb-CN01, little HMW was observed. Cot-duo-
cot did
not make HMWs for all four clones.
47
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
Example 3. Binding affinity analysis of antibody to mPDGFR-I3 and cotinine
(Enzyme immunoassays)
100 ng of mPDGFR-13-Fc chimera or TNF-a receptor extracellular domain-human Fc

fusion protein contained in a coating buffer (0.1 M sodium bicarbonate, pH
8.6) was coated on
a microtiter plate at 4 C 0/N. Each well was blocked with 150 I., of 3% (w/v)
BSA (bovine
serum albumin) in PBS as a blocking agent at 37 C for 1 hour. Various
concentrations (1:500-
1:62,500) of chicken serum diluted in 3% BSA/PBS was treated and incubated at
37 C for 2
hours. The microtiter plate was washed 3 times with 0.05% PBST, treated with
horseradish
peroxidase (HRP)-anti chicken IgY antibody (Millipore, Billerica, MA, USA),
and incubated
at 37 C for 1 hour. The microtiter plate was once again washed with 0.05%
PBST, and then
cultured using 2,T-azino-bis-3-ethylbenzothiazoline-6-sulfonic acid solutions
(ABTS) (Pierce,
Rockford, IL, USA). Then, the absorbance was measured at 405 nm with a
Multiscan Ascent
microplate instrument (Labsystems, Helsinki, Finland).
100 ng of mPDGFR-13-Fc chimera in the coating buffer or extracellular region
of TNF-
a receptor-human Fc fusion protein was coated on a microtiter plate at 4 C
0/N. Each well was
blocked with 150 I., of 3% (w/v) BSA (bovine serum albumin) in PBS as a
blocking agent at
37 C for 1 hour. After treatment with various concentrations of bispecific
antibodies (0.06 nM-
1 p,M) diluted in 3% BSA/PBS, they were incubated at 37 C for 2 hours. The
plate was washed
3 times with PBST and incubated for 1 hour at 37 C using HRP-anti-human CI<
antibody
(Millipore). After washing the plate 3 times with PBST, 3,3 ',5,5'-tetramethyl
benzidine
substrate solution (TMB) (GenDEPOT, Barker, TX, USA) was reacted. Then, the
absorbance
was measured at 650 nm with a Multiscan Ascent microplate instrument
(Labsystems).
48
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
In order to confirm the competition between the bispecific antibody according
to the
present invention and mPDGF-BB, the microtiter plate was coated with mPDGFR-13-
Fc
chimera as mentioned above and then blocked. The microtiter plate was treated
with various
concentrations of the bispecific antibody (0.06 nM-1 pM) with or without
addition of mPDGF-
BB (100 nM), and then incubated at 37 C for 2 hours. The washing, incubation,
and detection
were performed in the same manner as the enzyme immunoassay described above.
Enzyme immunoassay was performed to confirm the simultaneous binding ability
of
the bispecific antibody of scFv-C-K-scFy fusion protein to mPDGFR-13 and
cotinine. The four
kinds of bispecific antibodies did not bind to the control-human Fc protein
(Fig. 3b), but bound
to mPDGFR-13-Fc chimeric protein in a concentration-dependent manner (Fig.
3a).
The bispecific antibody containing the anti-mPDGFR-13 antibody according to
the
present invention had a binding ability to cotinine-BSA, but no binding
ability was observed
in the case of anti-HER2 x cotinine scFv-C-K-scFy fusion protein which was
used as a control
bispecific antibody (Fig. 3c).
In order to confirm whether the bispecific antibody has simultaneous binding
ability
to both cotinine and mPDGFR-13, anti-mPDGFR-13 x cotinine scFv-C-K-scFy fusion
protein was
reacted in wells coated with cotinine-BSA. Washing was performed for each
process, and
mPDGFR-13-Fc chimeric protein and HRP-anti human Fc antibody were sequentially
incubated.
Unlike other control groups, the four types of bispecific antibodies of scFv-C-
K-scFy (PRb-
CC01, PRb-CN01, PRb-CCO2, PRb-CC03) simultaneously bound to mPDGFR-13 and
cotinine
(FIG. 3d).
A competition enzyme immunoassay was developed to examine the effect of mPDGF-
BB on the binding ability of anti-mPDGFR-13 x cotinine scFv-C-K-scFy fusion
protein. The
49
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
serially diluted bispecific antibodies (anti-mPDGFR-13 scFv-C-k-cotinine scFv)
were incubated
on wells coated with mPDGFR-13-Fc chimeric fusion protein with or without
mPDGF-BB.
HRP-anti-human CI< antibody was incubated and then TMB was added. The binding
ability of
PRb-CC01, PRb-CCO2, and PRb-CCO3 to mPDGFR-13 was inhibited in the presence of

mPDGF-BB (Fig. 4a). However, the binding ability of PRb-CN01 was not
inhibited, so it
confirmed that PRb-CN01 binds to mPDGFR-13 in a noncompetitive manner to mPDGF-
BB.
Example 4. Internalization analysis of antibody (confocal microscopy analysis)
The bispecific antibody of scFv-C-k-scFv fusion protein was analyzed with a
confocal
microscope for visualizing internalization. NIH3T3 cells were treated with the
bispecific
antibody of scFv-Cic-scFv (10 p.g/mL) diluted in DMEM containing 10% FBS, and
internalized
at 37 C for 30 minutes. After washing the cells 3 times with cold PBS, acidic
buffer (0.2 M
acetic acid, 0.5 M sodium chloride) was treated at room temperature for 5
minutes to remove
the antibody bound to the cell surface. Cells were washed twice with cold PBS,
fixed with 4%
paraformaldehyde for 10 minutes, and then subjected to immunofluorescence
staining as
previously described [J.M. Lim et al., J. Cell. Biol. 210 (2015) 231.
Briefly, to block antibodies having non-specific reaction, cells were
incubated in PBS
containing 5% horse serum and 0.1% Triton X-100 for 30 minutes, and 2 pg/mL
FITC-anti
human CI< antibody (TB28 -2, BD Biosciences) was added at room temperature for
30 minutes.
To image the initial endosomes, the cells were washed 3 times with PBS,
incubated with PBS
containing 5% horse serum and 0.1% Triton X-100 for 30 minutes, and then
incubated with
ati-Rab5 antibody diluted to 1:200 (C8B1, Cell Signaling Technology, Danvers,
MA, USA).
Alexa Fluor 546 goat anti-rabbit IgG (A-11035, Invitrogen) was sequentially
treated. Cells
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
were treated with 0.2 pg/mL DAPI to detect DNA. Confocal microscopy images
were obtained
using a Zeiss LSM 880 microscope, and images were analyzed with Zen software
(Carl Zeiss,
Thornwood, NY, USA).
It was confirmed that three types of anti-mPDGFR-13 x cotinine scFv-Cx-scFv
fusion
proteins were internalized to cells through endosomes.
To measure the cellular internalization of the bispecific antibody of scFv-Cx-
scFv,
anti-mPDGFR-13 x cotinine scFv-Cx-scFv fusion protein was incubated in NIH3T3
cells, and
then treated with FITC-anti-human CI< antibody and endosome-specific antibody.
It was
imaged using a confocal microscope. Intracellular fluorescence was detected
fro only in cells
cultured with PRb-CN01, PRb-CCO2, and PRb-CCO3 (Fig. 4c). PRb-CC01 was not
internalized. When the images were merged, the present inventors confirmed
that the
fluorescence of the three antibody clones and the endosome-specific antibody
were co-
localized.
Example 5. Binding ability analysis of a bispecific antibody to mPDGFR-I3 and
cotinine (flow cytometry analysis)
NIH3T3 cells were incubated with chicken serum diluted 1:100 in flow cytometry

buffer (1% [w/v1 BSA/PBS in 0.05% [w/v1 sodium azide) at 4 C for 1 hour and
washed twice
with flow cytometry buffer. The cells were treated with Alexa Fluor 488-anti
chicken IgY
antibody (703-545-155, Jackson Immunoresearch, West Grove, PA, USA) in flow
cytometry
buffer. After washing, it was classified and analyzed with a FACS Canto II
instrument (BD
Biosciences, San Jose, CA, USA). 10,000 cells were used for each measurement,
and the results
were analyzed by FlowJo (Tree Star, Ashland, OR, USA). The NIH3T3 cells were
prepared by
51
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
being purchased from Korea Cell Line Bank and incubated in Dulbecco's modified
Eagle's
medium (DMEM; Welgene, Seoul, Korea) with added by 10% fetal bovine serum
(FBS;
GIBCO, Grand Island, NY, USA), 1% penicillin, and streptomycin [S. Park et
el., Exp. Mol.
Med. 44 (2012) 554-61].
mPDGF-BB was biotinylated using a biotin-xx microscale protein labeling kit
(Invitrogen) according to the manufacturer's instructions. NIH3T3 cells were
cultured at 4 C
for 1 hour using the bispecific antibody of scFv-Cx-scFv fusion protein (100
nM) under two
different conditions with or without mPDGF-BB-biotin (100 nM). The cells were
washed 4
times with flow cytometry buffer and cultured with allophycocyanin (APC)-anti-
human CI<
antibody (clone TB28-2; BD Biosciences, San Jose, CA, USA) and streptavidin-
phycoerythrin
(PE) (12-4317-87; eBioscience, ThermoFisher). After washing, it was classified
and analyzed
with a FACS Canto II instrument (BD Biosciences, San Jose, CA, USA). 10,000
cells were
used for each measurement, and the results were analyzed by FlowJo (Tree Star,
Ashland, OR,
USA). Like the result from the enzyme immunoassay method, only PRb-CN01 was
bound to
PDGFR-f3 present on the cell surface (FIG. 4B), when mPDGF-BB was added (FIG.
4b). The
binding ability of the other three clones was inhibited by mPDGF-BB.
The present inventors tested the binding ability of the bispecific antibody of
scFv-Cx-
scFv fusion proteins in MOLT-4 cells not expressing mPDGFR-13 by flow
cytometry analysis.
MOLT-4 cells were cultured with a bispecific antibody of scFv-Cx-scFv fusion
protein (100
nM) at 4 C for 1 hour. After washing as described above, the detection was
conducted by
treating with an APC-anti human CI< antibody. The MOLT-4 cells were purchased
from the
Korea Cell Line Bank and prepared by culturing in RPMI-1640 (Welgene, Seoul,
Korea)
medium added by 10% fetal bovine serum, 1% penicillin, and streptomycin. It
was confirmed
52
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
that neither PRb-CNO1 nor the anti-HER2 bispecific antibody of scFv-Cx-scFv
fusion antibody
binds to the corresponding cells (Fig. 6).
Example 6. Complex Formation of bispecific antibody and cotinine-duocarmycin
(cot-duo)
6-1: cotinine-duocarmycin conjugate synthesis
The present invention prepared valine-citrulline-PAB-duocarmycin, valine-
citrulline-
PAB-monomethyl auristatin E (MMAE) or valine-citrulline-PAB-mailimidomethyl
cyclohexane-l-carboxial Rate (mcc) mertansine (DM1) which were conjugated with
cotinine.
In the present invention, as a result of testing the cotinine-cytotoxic drugs
of DAR1 and DAR4,
it was confirmed that the DAR4 cotinine-cytotoxic drug was more potent than
DAR1, and also
found that duocarmycin was the most potent when bound to cotinine. For this
reason, cotinine-
duocarmycin was used in the experiment.
Trans-4-cotinine carbonyl-(GSK)4 peptide was synthesized by Fmoc solid phase
peptide synthesis 9SPSS using ASP48S auto peptide synthesizer in Peptron
(Daejeon, Korea).
Trans-4-cotinine carboxylic acid (Sigma-Aldrich, St Louis, MO, USA) was
attached to the N-
terminus of the peptide using the Fmoc-amino acid coupling method.
After the synthesis was completed, the crude product was separated from the
resin by
treating TFA/EDT/thioanisole/TIS/DW (90/2.5/2.5/2,5/2.5 volume) for 2 hours.
The solution
was precipitated by performing centrifugation using cold ether. The
precipitates were air dried.
The crude product was purified by reverse phase HPLC using an ACE 10 C18-300
reverse
phase column (250 mm x 21.2 mm, 10 04). It was eluted with a water-
acetonitrile linear
53
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
gradient (10-75% (v/v) acetonitrile) containing 0.1% (v/v) trifluoroacetic
acid (Alfa Aesar,
Warm Hill, MA, USA). The purified peptide (Cot-(GSK)4 peptide) was collected
and dried.
Valine-citrulline p-aminobenzyloxycarbonyl (PAB)-linked dimethylaminoethyl
duocarmycin was linked to free four amino acids of lysine in cotinine-(GSK)4
by Levena
Biopharma (San Diego, CA, USA). Cot-(GSK)4 peptide (3.5 mg, 2 mol) was
dissolved in
acetonitrile/water (6/4, v/v, 1 mL). NHS ester of PAB-dimethylaminoethyl
duocarmycin
PEG3-valine-citrulline was added, and 9 1., of saturated aqueous NaHCO3 was
sequentially
added. The mixture was mixed at room temperature for 4 hours, and purified by
reverse phase
HPLC technique using a Phenomenex Gemini C18-100A column (100 mm x 2 mm x 5
M).
The complex (cotinine-[GSK(duocarmycin)]4. DAR4) was designated as -cot-duo".
Bivalent-
cotinine-(GSK)4K was linked to duocarmycin by the method described previously
[J. Jin et al.,
Exp. Mol. Med. 50 (2018) 671. Briefly, two trans-4-cotinine carboxylic acid
molecules from
Peptron were linked to the free amino acid at the N-terminus of GSKGSKGSKGSKK
and the
epsilon amino acid at the C-terminus of lysine using a basic Fmoc-amino acid
coupling method.
In Levena Biopharma, four PAB-duocarmycins were linked with bivalent cotinine-
GSKGSKGSKGSKK peptides to form a complex named cotinine-[GSK(duocarmycin)]4K-
cotinine (DAR2) or cot-duo-cot. Fig. la shows the fusion protein of the
bispecific antibody and
cotinine-duocarmycin conjugate (cot-duo, cot-duo-cot), and Fig. lb shows the
chemical
structures of cot-duo and cot-duo-cot. ``R" is yaline-citrulline PAB-linked
dimethyl aminoethyl
duocarmycin.
6-2: Complex formation with a bispecific antibody
54
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
Anti-PDGFR-13 x cotinine scFy-C-K-scFy fusion protein (15 0/1) dissolved in
PBS
obtained in Example 2 was mixed at a ratio of 1:1 with cot-duo (15 0/1)
dissolved in DMSO,
and mixed at a ratio of 2:1 with cot-duo-cot (7.5 0/1). After 30 minutes of
complex formation
at room temperature, the complexes were diluted 5-fold in DMEM medium
containing 10%
FBS and 1% penicillin/streptomycin (25.6 pM-2 0/1).
6-3: Confirmation of complex formation with a bispecific antibody
To confirm the complex formation, the (anti-mPDGFR-13 scR)-C-K-(anti-cotinine
scFv)
of bispecific antibody and cotinine-duocarmycin conjugates prepared in Example
2 were
analyzed by Y-Biologics (Daejeon, Korea) using size exclusion chromatography
and HPLC.
For the analysis, a Dionex Ultimate 3000 (Thermo Fisher Scientific Inc., MA,
USA)
equipped with a Sepax SRT-C SEC-300 column (7.8 x 300 mm) filled with 300 A-
sized pores
in 5 p.m particles was used. The mobile phase was PBS (phosphate-buffered
saline), and 20 pt
of a sample (1 mg/mL) was injected, and the eluate was eluted at 1 mL/min for
15 minutes.
The eluate from the column was monitored with an ultraviolet detector at 254
nm with mAU
values.
Fig. 8 is a result of size exclusion chromatography of a bispecific antibody
containing
an anti-mPDGFR-13 antibody according to the present invention. The bispecific
antibodies, the
complex of bispecific antibodies and cot-duo, the complex of bispecific
antibodies and cot-
duo-cot according to the present invention were analyzed with the size
exclusion
chromatography-HPLC using Dionex Ultimate 3000 equipped with a Sepax SRT-C SEC-
300
column. The mobile phase was used as PBS, and the mobile solvent was eluted at
1 mL/min
for 15 minutes. The ultraviolet detector was adjusted to 254 nm and the
results were monitored
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
by mAU. HMW is a high molecular weight species.
Example 7. Anti-proliferative analysis in PDGFR-13-expressing cell lines
(cytotoxicity test)
NIH3T3 cells were added to 50 !IL of DMEM supplemented with 10% FBS and 1%
penicillin/streptomycin, placed in a 96-well plate (CLS3595, Corning) and
incubated overnight
in 5% CO2 at 37 C incubator. In each well containing 50 !IL of cells, 50 !IL
of 50 anti-PDGFR-
(3 x cotinine scFv-Cx-scFy fusion protein (25.6 pM-2 04) complexed with cot-
duo or cot-duo-
cot obtained in Experimental Example 10 was added and incubated for 72 hours
in 5% CO2 at
37 C incubator. To test mPDGF-BB on the toxic effect of the bispecific
antibody of scFv-Cx-
scFy and the cotinine-duocarmycin complex, mPDGF-BB (2 nM) was added together
with 50
!IL of the bispecific antibody scFv-Cx-scFy and the cotinine-duocarmycin
complex. The mixed
complexes were added to 50 !IL of NIH3T3 cells.
The bispecific antibody of anti-HER2 x cotinine scFv-Cx-scFy was used as a
control.
After 72 hours of incubation in the cells, 100 !IL of Cell Titer-Glo reagents
(Promega Corp.,
Madison, WI, USA) were added to all wells according to the manufacturer's
instructions, and
luminescence was measured in a luminometer (PerkinElmer, Waltham, MA, USA).
The
experiment was repeated 3 times. Relative viability was calculated with the
following formula:
[% viability = (luminescence of experimental well-luminescence of background
well)/
(luminescence of control-luminescence of background well) x 1001. The wells
containing only
new medium were used as background wells.
56
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
It was confirmed that the complex of anti-mPDGFR-13 x cotinine scFv-C-K-scFv
fusion
antibody and cotinine duocarmycin (cot-duo, cot-duo-cot) had antiproliferative
effects on
mouse fibroblasts expressing PDGFR-13.
To evaluate the toxicity of the complex of anti-mPDGFR-B x cotinine scFv-C-K-
scFv
bispecific antibodies and cotinine duocarmycin (cot-duo, cot-duo-cot), the
complexes were
added to NIH3T3 cells in the presence/absence of mPDGF-BB. The ATP of the
cells was
measured for relative cell viability. The toxicity of the bispecific antibody
of scFv-C-K-scFv
fusion antibody was shown by IC50 in Table 6 and Fig. 5. Table 6 below shows
IC50 of the in
vitro titer and 95% confidence interval of the complex of the anti-mPDGFR-13 x
cotinine scFv-
C-K-scFv fusion antibody cotinine duocarmycin.
[Table 6]
Cot-duo Cot-duo-cot
Bispecifi anti-mPDGFRO x anti-cotinine scFv-Cx IC50 (nM) 95% CI IC50
(nM) 95% CI
PRIa-CCO1
with mPDGF-BB 27.1 19.1 - 38.5 67.2 52.0- 86.9
without mPDGF-BB 25.0 18.1 - 34.7 87.6 54.4- 141.1
PRP-CHOI
with mPDGF-BB 9.7 8.4 - 11.1 30.5 25.1- 37.1
without mPDGF-BB 6.3 5.1 - 7.9 20.1 138-29.4
PRIa-CCO2
with mPDGF-BB 20:6 16.2- 26.3 69.0 54.2- 87.9
without mPDGF-BB 20.1 15.2 - 26.5 125.4 47.4 - 328.7
PRID-CCO3
with mPDGF-BB 20.7 15.7- 27.3 50.2 39.5- 63.9
without mPDGF-BB 27.3 8.9- 84.3 67.3 39.0-1162
Anti-HER2
with mPDGF-BB 23.2 17.4- 31.0 60.8 484- 76.4
without mPDGF-BB 36.7 27.9-48.3 93.7 55.3-158.9
Cotinine duocarmycin conjugates
with mPDGF-BB 22.8 16.6- 31.4 56.7 443- 72.6
without mPDGF-BB 15.5 11.7 - 20.3 45.4 298-69.2
Free duocarmycin
with mPDGF-BB 23.8 16.2- 31.0 60.3 41.1- 88.5
without mPDGF-BB 9.7 7.3- 12.9 35.7 234- 54.4
Of the four tested antibodies, PRb-CNO1 showed the highest toxicity with or
without
mPDGF-BB, compared to the control group of anti-HER2 x cotinine scFv-C-K-scFv
(p<0.01;
Fig. 5). PRb-CCO2 and PRb-CCO3 showed toxicity compared to the control
antibody and the
non-internalizing antibody (PRb-CC01), but did not show statistical
significance. However, in
57
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
the presence of mPDGF-BB, cotinine-duocarmycin was significantly more toxic
than free
duocarmycin, when complexed with PRb-CN01 (p < 0.01). As a control experiment,
the
toxicity experiments were performed on MOLT-4 cells that do not express mPDGFR-
13 with
the bispecific antibody of scFv-Cx-scFy fusion protein and cot-duo complex.
PRb-CNO1 had
no difference in toxicity from the control antibody of anti-HER2 x cotinine
scFv-Cx-scFy
fusion protein (FIG. 7).
As shown in Fig. 5, scFv-Ck-scFy did not kill the cells even when the antibody

concentration increases, so the IC50 value cannot be calculated. In table 6,
the result of addition
of cot-duo or cot-duo-cot together was indicated instead of IC50 of scFv-Ck-
scFy alone. In
addition, since MOLT-4 cells do not express mPDGFR-13, there is no difference
in toxicity
between the PRb-CNO1-drug complex and the control antibody-drug complex.
Example 8. Analysis of neovascularization in vivo
It was confirmed that the complex of anti-mPDGFR-13 x cotinine scFv-Cx-scFy
fusion
antibody conjugated with cot-duo inhibited neovascularization in vivo.
8-1: Experiment of animal with oxygen-induced retinal disease
C57BL/6 mice on 7th day after birth were reared in a high-concentration oxygen
(75%)
chamber for 5 days, and transferred to a normal oxygen breeding facility on
12th day to create
an environment for retinopathy of premature baby.
On the 14th day after birth, 1 uL of each bispecific antibody scFv-Cx-scFv,
and the
complex of the bispecific antibody scFv-Cx-scFy fusion protein and cot-duo
were injected into
58
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
the vitreous cavity at a concentration of 1 nM and 10 nM, respectively. To
label blood vessels,
isolectin B4-594 (1:100; 121413, Invitrogen) was treated on the retinal flat
mount.
In an oxygen-induced retinal disease animal model, the complex of the PRb-CN01
x
cotinine scFv-C-K-scFv fusion antibody cot-duo inhibited neovascularization in
a
concentration-dependent manner (FIG. 9).
8-2: Laser-induced choroidal neovascularization animal model
After anesthetizing the mice, the retina of the mouse was irradiated with an
indirect
ophthalmoscope system (ILOODA) laser in a 300 um spot size with 400 mW
intensity, 50 ms
duration, and 810 nm wavelength to induce destruction of Bruch's membrane.
After 4 days of laser irradiation, 1 uL of each bispecific antibody scFv-C-K-
scFv, and
complex of the bispecific antibody scFv-C-K-scFv fusion protein and the cot-
duo was injected
into the vitreous cavity at a concentration of 10 nM. After 7 days of laser
irradiation, the eyes
were removed to obtain retinal pigment epithelial-choroid-scleral tissue, and
immunofluorescence staining was performed with isolectin B4 antibody
conjugated to Alexa
Fluor 594 to determine the extent of the choroidal neovascular membrane. The
neovascularization inhibition ability of the scFv-C-K-scFv fusion protein and
the cot-duo
complex was confirmed. To quantitatively analyze the extent of the choroidal
neovascular
membrane, the ImageJ program (NIH) was used.
In the laser-induced choroidal neovascularization animal model, the PRb-CNO1 x

cotinine scFv-C-K-scFv fusion antibody cot-duo complex inhibited
neovascularization (FIG. 10).
The toxicity of the bispecific antibody scFv-C-K-scFv was calculated as IC50
(50% of
the maximum inhibitory ability), and statistics analysis were performed by
unpaired Student's
59
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
t-tests or one-way analysis of variance. Tukey's post hoc multiple comparison
test was used to
see the statistical significance of the bispecific antibodies scFv-Cx-scFv. P-
value of 0.05 or
less was considered statistically significant. All analyzes were performed
using Prism v5.0
(GraphPad Software, Inc., San Diego, CA, USA).
Example 9. Expression and purification of hPDGFR-13-Cx fusion protein
An amino acid sequences for the extracellular regions of hPDGFR-13, Cynomolgus

monkey PDGFR-13, and Sus scrofa PDGFR-P are shown in SEQ ID NOs: 61, 62, and
63 in
Table 3 above, but rhesus PDGFR-13 is commercially available and purchased as
the antigens
(Cat: 90215-CO2H). The prepared nucleotide sequences (Ginscript Biotag,
Jangsuseong, China)
were synthesized with addition of SfiI restriction recognition sequence at the
end of the
prepared nucleotide sequence, digested with Sfi/ enzyme, and cloned into pCEP4
vector. Then,
the proteins were expressed in the form of CI< or hFc according to the
previously reported
method [Y. Lee, H. Kim et al., Exp. Mol. Med. 46 (2014) e114, S. Park, D. Lee
et al., Clin
Chim Acta 411 (2010) 12381.
The proteins were transfected and purified according to the same method of
mPDGFR-
(3 in Example 1, but modified to purify the hFc fusion protein using Protein A
gel affinity
chromatography according to the manufacturer's instructions (Repligen Corp.,
Cambridge,
MA). The fusion protein has a structure in which CI< is linked to the C -
terminus of an amino
acid sequence hPDGFR-13 through a linker-(GGGGS)3.
Example 10. Expression and purification of bispecific antibodies (scFv-Cx-
scFv)
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
10-1: Preparation of combinatorial scFv-displayed phage library and bio-
panning
according to the same method of Preparation of combinatorial scFv-displayed
phage library
and bio-panning in Example 2, an anti-hPDGFR-13 scFv-expressing phage library
was produced
using the hPDGFR-[3-Cic fusion protein in Example 9 and was performed
biopanning.
At the 5th output titer, scFv clones were randomly selected and phage enzyme
immunoassay was performed on mPDGFR-13-C-k-coated microtiter plates. After
analyzing an
amino acid sequence, all 3 types of antibody clones were identified (Table 7).
Table 7 below
shows the H-CDR3 sequences of anti-hPDGFR-13 scFv clones. Table 2 shows the VH
and VL
and their CDR amino acid sequence information for the obtained three clones.
[Table 7]
Clone Amino acid sequence of VH-CDR3 SEQ ID NO
PRb-CN16 AAGTCYSHSCTGYIDA 35
PRb-CN32 SAGSTY SY WD SDAGLIDA 43
PRb-CN26 RGFMDAGGIDA 51
10-2: Expression and purification of a bispecific antibody scFv-Cx-scFv fusion

protein
Using the anti-hPDGFR-13 scFv clone prepared in Example 9, (anti-hPDGFR-f3
scFv)-
C-k-(anti-cotinine scFv) A pCEP4 expression vector containing a gene encoding
a bispecific
antibody was prepared according to the same method as the preparation of anti-
mPDGFR-13
scFv in Example 2.
The specific structure of the prepared (anti-hPDGFR-13 scFv)-C-k-(anti-
cotinine scFv)
bispecific antibody and the binding positions of each linker are shown in FIG.
la. Specifically,
the anti-hPDGFR-13 scFv and anti-cotinine scFv are each joined in the order of
VL-
GQSSRSSGGGGSSGGGGS (SEQ ID NO: 57 in Table 2)-VH from the N terminal, and that
61
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
is, the C terminal of VL and the N terminal of VH are connected. From the N-
terminus, anti-
hPDGFR-f3 scFv, Ck and anti-cotinine scFv are linked using each linker, and
the linker
connecting the scFv is (GGGGS)3 of SEQ ID NO: 59 in Table 2, and Ck amino
acids is shown
in SEQ ID NO: 58 in Table 2.
The specific method of preparing a fusion protein of a bispecific antibody,
the
purification and the purity measurement of the expressed bispecific antibodies
were performed
in the substantially same method of Example 2. The result of SDS-
polyacrylamide gel
electrophoresis is shown in Fig. 11. Fig. 11 is a result of SDS-polyacrylamide
gel
electrophoresis of a cotinine scFv-C-K-scFv fusion protein as a bispecific
antibody against the
anti-hPDGFR-13 according to the present invention.
As shown in Fig. 11, the protein reduced using a reducing agent at lane 1 (PRb-
CN16
x cotinine bispecific antibody), lane 3 (PRb-CN26 x cotinine bispecific
antibody) and lane 5
(PRb-CN32 x cotinine bispecific antibody) was stained at 69.0 kDa, the reduced
protein was
stained at 67 kDa, and the non-reducing protein was stained at 60 kDa. The
computer-
calculated fusion protein size was 66.77 kDa. No multimeric bands were
observed.
Example 11. Binding affinity analysis of bispecific antibodies to hPDGFR-13
and
cotinine
100 ng of hPDGFR-13-Fc chimera or TNF-a receptor extracellular domain-human Fc

fusion protein contained in a coating buffer (0.1 M sodium bicarbonate, pH
8.6) was coated on
a microtiter plate at 4 C 0/N. Each well was blocked with 150 jiL of 3% (w/v)
BSA (bovine
serum albumin) in PBS as a blocking agent at 37 C for 1 hour. Various
concentrations (1:500-
1:62,500) of chicken serum diluted in 3% BSA/PBS was treated and incubated at
37 C for 2
62
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
hours, and analyzed according to the same method of the protocol for testing
the anti-
mPDGFR-13 immune chicken serum binding ability.
100 ng of hPDGFR-13-Fc chimera in the coating buffer or extracellular region
of TNF-
a receptor-human Fc fusion protein was coated on a microtiter plate at 4 C
0/N. Each well was
blocked with 150 j.tL of 3% (w/v) BSA (bovine serum albumin) in PBS as a
blocking agent at
37 C for 1 hour. After treatment with various concentrations of bispecific
antibodies of scFv-
Cx-scFy fusion protein (0.01 nM- 1 p,M), the analysis was performed in the
same protocol of
the concentration-dependent binding ability of anti-mPDGFR-13 scFv-Cx-scFy
fusion protein.
In order to confirm the competition between the bispecific antibody scFv-Cx-
scFy and
hPDGF-BB, as in Example 3, hPDGFR-13-Fc chimera was coated on a microtiter
plate and then
blocked. The microtiter plate was treated with various concentrations of the
bispecific antibody
(0.01 nM-1 p,M) with or without addition of hPDGF-BB (100 lily!; 220-BB; R&D
systems),
and then incubated at 37 C for 2 hours. The washing, incubation, and detection
were performed
in the same manner as the enzyme immunoassay described above. The binding
ability of PRb-
CN16, PRb-CN32, and PRb-CN26 to hPDGFR-13 was not inhibited in the presence of
hPDGF-
BB (Fig. 12a).
100 ng of hPDGFR-13, mPDGFR-13, rhesus PDGFR-13 (90215-CO2H, Sino Biological
Inc. Beijing, China), cynomolgus monkey PDGFR-13, Sus scrofa PDGFR-P-Fc
chimera or
TNF-a receptor extracellular domain-human Fc fusion protein was coated with 4
C
temperature 0/N. After blocking the wells, sequential culture of the anti-
hPDGFR-13 scFy-Cx-
scFy fusion protein and the HRP conjugated anti-human CI< antibody was
followed in the same
manner as in Example 3.
63
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
Enzyme immunoassay was performed to confirm the simultaneous binding ability
of
the bispecific antibody of scFv-C-K-scFy fusion protein to hPDGFR-13 and
cotinine. The four
kinds of bispecific antibodies did not bind to the control-human Fc protein
(Fig. 12b), but
bound to hPDGFR-13-Fc chimeric protein in a concentration-dependent manner
(Fig. 12a).
Both the anti-hPDGFR-13 x cotinine scFv-C-K-scFy fusion protein and the
control
bispecific antibody of anti-mCD154 x cotinine scFv-C-K-scFy fusion protein had
the binding
ability to cotinine-BSA (Fig. 12c). In order to determine of the simultaneous
binding ability of
cotinine and hPDGFR-13, an anti-hPDGFR-13 x cotinine scFv-C-K-scFy fusion
protein was
cultured in wells coated with cotinine-BSA. Washing was performed for each
process, and
hPDGFR-13-Fc chimeric protein and HRP-anti human Fc antibody were incubated in
order. The
three kinds of bispecific antibody scFv-C-K-scFy fusion proteins
simultaneously bound to
hPDGFR-13 and cotinine (Fig. 12d).
It was confirmed that the anti -hPDGFR-f3 x cotinine scFv-C-K-scFy fusion
protein
could simultaneously bind to hPDGFR-13 and cotinine even in the presence of
hPDGF-BB (Fig.
13a).
Enzyme immunoassay was used to test the cross-species binding of the anti-
hPDGFR-
(3 x cotinine scFv-C-K-scFy fusion protein. All three clones except the
control x cotinine scFv-
C-K-scFy fusion protein successfully bound to hPDGFR-13, mPDGFR-13, Rhesus
PDGFR-13,
Cynomolgus monkey PDGFR-13, and Sus scrofa PDGFR-13 (Fig. 17).
Fig. 17 is a graph showing an experiment of the cross-species reaction of the
bispecific
antibody against the anti-hPDGFR-13 antibody according to the present
invention. On the
microtiter plates coated with hPDGFR-13, mPDGFR-13, Rhesus PDGFR-13,
Cynomolgus
monkey PDGFR-13, Sus scrofa PDGFR-13 and control Fc coated, the bispecific
antibody
64
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
containing anti-hPDGFR-fl antibody according to the present invention (100 nM)
was reacted
and the amount of bound bispecific antibody was measured using HRP conjugated
anti-human
CI< antibody and TMB.
Example 12. Internalization analysis of bispecific antibody
Human pericyte cells were treated with the diluted bispecific antibody scFv-Cx-
scFv
(10 pg/mL) in pericyte growth medium supplemented with 10% FBS, and
internalized at 37 C
for 30 minutes. The analysis was performed in the same manner according to the
protocol of
the confocal microscope of the anti-mPDGFR-r3 x cotinine scFv-Cx-scFv fusion
protein of
Example 4. After the addition of hPDGF-BB (50 ng/mL) to the bispecific
antibody scFv-Cx-
scFv fusion protein, the confocal microscopy experiments were performed
according to the
same protocol.
It was confirmed that the anti-hPDGFR-fl x cotinine scFv-Cx-scFv fusion
protein was
internalized to cells through endosomes.
The internalization of the bispecific antibody scFv-Cx-scFv fusion protein was

visualized. After treatment of human pericyte cells with the fusion protein,
antibodies bound
to the cell surface were removed. After cell fixation, the fusion protein was
stained with FITC-
anti human CI< antibody (green). To visualize early endosomes, the cells were
stained with anti-
Rab5 antibody and Alexa Fluor 546-goat anti-rabbit antibody IgG (red). The
part indicated by
the arrow shows the enlarged part in which the fluorescence of the anti-hPDGFR-
fl x cotinine
scFv-Cx-scFv fusion protein and the initial endosome were co-localized. DNA
was stained
with DAPI (blue)(Scale bar, 10 pm). Fig. 13d was repeated with hPDGF-BB.
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
To measure the cellular internalization of the bispecific antibody of scFv-Cic-
scFv,
anti-hPDGFR-13 x cotinine scFy-Cic-scFv fusion protein was incubated in human
pericyte cells,
and then treated with FITC-anti-human CI< antibody and endosome-specific
antibody. It was
imaged using a confocal microscope. Intracellular fluorescence was detected
from only in cells
cultured with PRb-CN32 and PRb-CN16 (Fig. 13c). When the images were merged,
the present
inventors confirmed that the fluorescence of the two antibody clones and the
endosome-
specific antibody were co-localized. When PRb-CN32, PRb-CN16, and PRb-CN26
were
treated with hPDGF-BB, they were co-localized with endosome-specific antibody
(Fig. 13d).
PRb-CN26 was internalized in a large amount within 30 minutes when PDGF-BB was
treated
together. Internalization of the antibody is very important for delivery of
drug into the target
cell. In case that the level of PDGF-BB was elevated in some diseases, even
increased the level
of PDGF-BB did not affect the internalization of the antibody according to the
present
invention. Therefore, it is useful for intracellular delivery of drug to
patients with diseases.
Example 13. Binding ability analysis of a bispecific antibody to hPDGFR-13 and

cotinine
Human pericyte cells were incubated with chicken serum diluted 1:100 in flow
cytometry buffer (1% [w/v] BSA/PBS in 0.05% [w/v] sodium azide) at 4 C for 1
hour and
washed four times with flow cytometry buffer. The experiment followed the same
protocol of
that testing anti-mPDGFR-13 in Example 4, The human percyte cells were
prepared by being
purchased from PromoCell (Heidelberg, Germany) and cultured in Dulbecco's
modified
Eagle's medium (DMEM; Welgene, Seoul, Korea) supplemented with 10% fetal
bovine serum
66
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
(FBS; GIBCO, Grand Island, NY, USA), 1% penicillin, and streptomycin [S. Park
et el., Exp.
Mol. Med. 44 (2012) 554-61].
Human pericyte cells were cultured at 4 C for 1 hour using the bispecific
antibody of
scFv-Cic-scFv fusion protein (100 lily!) under two different conditions with
or without hPDGF-
BB-biotin (100 nM, BT220; R&D systems). The cells were washed 4 times with
flow
cytometry buffer and cultured with allophycocyanin (APC)-anti-human CI<
antibody (clone
TB28-2; BD Biosciences, San Jose, CA, USA) and streptavidin-phycoerythrin (PE)
(12-4317-
87; eBioscience, ThermoFisher). After washing, it was classified and analyzed
with a FACS
Canto II instrument (BD Biosciences, San Jose, CA, USA). 10,000 cells were
used for each
measurement, and the results were analyzed by FlowJo (Tree Star, Ashland, OR,
USA).
The bispecific antibody scFv-Cic-scFv fusion protein (100 nM) was incubated at
4 C
for 1 hour in A-431 cells. After washing as described above, the detection was
conducted by
treatment with an APC-anti human CI< antibody. The A-431 cells were purchased
from the
Korea Cell Line Bank and cultured in Dulbecco's modified Eagle's medium (DMEM;
Welgene,
Seoul, Korea) medium containing 10% fetal bovine serum (GIBCO), 1% penicillin,
and
streptomycin.
The present inventors tested the binding ability of each clone in human
pericyte cells
in the presence of hPDGF-BB by flow cytometry analysis, and confirmed that
anti-hPDGFR-
x cotinine scFv-Cx-scFy fusion protein and hPDGF-BB-biotin bound to the cells.
When
hPDGF-BB was added, all three clones were bound to PDGFR-fl present on the
cell surface,
which was similar to that of the enzyme immunoassay method (Fig. 13b). The
present inventors
confirmed the binding ability of the bispecific antibody scFv-Cx-scFy fusion
proteins to A-431
67
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
cells that do not express hPDGFR-13 by flow cytometry, and that all of the
anti-hPDGFR-13 x
cotinine scFv-Cx-scFv fusion proteins and the control bispecific antibody of
anti-mCD154 x
cotinine scFv-Cx-scFv fusion protein did not bind to the corresponding cells
(Fig. 15).
Example 14. Complex Formation of bispecific antibody and cotinine-
duocarmycin (cot-duo)
The experiment was performed in the substantially same method of Example 6,
except
that the bispecific antibody containing the anti-mPDGFR-13 antibody, a
bispecific antibody
related to the anti-hPDGFR-13 antibody prepared in Example 10 was used. As
described in the
preparation and transduction of the anti-mPDGFR-13 x cotinine scFv-Cx-scFv
fusion protein in
Example 6, HEI(293F cells were transduced in the same manner. The bispecific
antibody of
scFv-Cx-scFv fusion protein was purified from the supernatant by affinity
chromatography and
subjected to SDS-polyacrylamide gel electrophoresis (FIG. 11).
Example 15. Anti-proliferative analysis in PDGFR-13-expressing cell lines
(cytotoxicity test)
Human pericyte cells were added to 50 .1., of DMEM supplemented with 10% FBS
and 1% penicillin/streptomycin, placed in a 96-well plate (CLS3595, Coming)
and incubated
overnight in 5% CO2 at 37 C incubator. In each well containing 50 .1., of
cells, 50 .1., of 50
anti-hPDGFR-13 x cotinine scFv-Cx-scFv fusion protein (25.6 pM-2 04) complexed
with cot-
duo was added and incubated for 72 hours in 5% CO2 and 37 C incubator.
To evaluate the effect of hPDGF-BB on the toxicity of the complex of the
bispecific
antibody of scFv-Cx-scFv and cot-duo, hPDGF-BB (2 nM) was added together with
50 .1., of
68
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
the bispecific antibody scFv-C-K-scFv and cot-duo complex. The mixed complexes
were added
to 50 tL of cell-containing solution. The bispecific antibody anti-mCD154 x
cotinine scFv-
C-K-scFv was used as a control. Other experimental methods were carried out in
the same
manner as in the protocol of the anti-mPDGFR-13 x cotinine scFv-C-K-scFv
fusion protein in
Example 7.
In order to evaluate the toxicity of the anti-hPDGFR-f3 x cotinine scFv-C-K-
scFv fusion
protein and cot-duo complexes, the complex was treated in human pericyte cells
with or
without hPDGF-BB, and the cells were treated with adenosine triphosphate to
measure the
relative viability. The toxicity of the bispecific antibody of anti-hPDGFR-13
x cotinine scFv-
C-K-scFv fusion protein and cot-duo complex was shown by IC50 (Table 8). Table
8 below
showed in vitro titer with 95% confidence interval of the complex of the anti-
mPDGFR-13 x
cotinine scFv-C-K-scFv fusion antibody and cotinine duocarmycin.
[Table 8]
anti-hPDG FRO x anti-cotinine scFv-Cx-scFv Cot-duo
IC50 (nM) 95% CI
PRb-CN16
Without hPDGF-BB 2.39 2.12-2.71
With hPDGF-BB 1.82 1.58-2.10
PRb-CN32
Without hPDGF-BB 0.78 0.75-0.81
With hPDG F-BB 0.67 0.58-0.76
PRb-CN26
Without hPDGF-BB 0.90 0.84-0.98
With hPDG F-BB 0.87 0.78-0.96
Control
Without hPDGF-BB 4.47 6.73-13.87
With hPDGF-BB 3.48 3.05-3.97
Without hPDGF-BB 13.70 12.58-14.93
With hPDG F-BB 12.42 10.42-14.80
69
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
As shown in the results of Table 8, all of three tested antibodies (PRb-CN16,
PRb-
CN32, PRb-CN26) showed higher toxicity than control anti-mCD154 x cotinine
scFv-Cic-scFy
with or without hPDGF-BB (p < 0.05; Fig. 14a and Fig. 14b). As a control
experiment, the
toxicity experiments were performed on A-431 cells that did not express hPDGFR-
13, with the
bispecific antibody scFv-Cic-scFy fusion protein and cot-duo complex. All of
the anti-
hPDGFR-f3 x cotinine scFv-Cic-scFy fusion proteins had not higher toxicity
than the control
antibody anti-mCD154 x cotinine scFv-Cic-scFy fusion protein (FIG. 16).
Therefore, as a result
of anti-proliferative analysis result, the bispecific antibody/cot-duo complex
had higher
cytotoxicity in the human pericyte cell lines expressing hPDGFR-13, compared
to the control
anti-mCD154 x cotinine scFv-C-k-scFv, but there was no difference in
cytotoxicity in A-431
not expressing hPDGFR-13, compared to the control anti-mCD154 x cotinine scFv-
C-k-scFv.
This cytotoxicity result is caused by the efficacy of duocarmycin delivered by
hPDGFR-13,
which suggests the bispecific antibody scFv-Cic-scFy effectively delivers the
hapten-drug (cot-
duo) to target cells and effectively releases drugs intracellularly.
Example 16
First, using the PRb-CN01 scFy prepared in Example 2, a pCEP4 expression
vector
containing a gene encoding scFv-rabbit Fc antibody was constructed. An amino
acid sequence
of the rabbit Fc antibody used in the experiment is shown in SEQ ID NO: 72 in
the sequence
listing.
Specifically, it was linked to the rabbit Fc using a hinge (QEPKSSDKTHTSPPSP:
SEQ ID NO: 73) at the C-terminus of the anti-mPDGFR-13 scFv. As a specific
method for
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
preparing scFv-rabbit-Fc, the gene encoding anti-mPDGFR-13 was digested with
Sfi/ (New
England Biolabs) and ligated to an expression vector. Transfection and
purification were
performed according to the method for mPDGFR-13 in the Examples, but it was
modified to
purify the rabbit Fc fusion protein using Protein A gel affinity
chromatography according to
the manufacturer's instructions (Repligen Corp., Cambridge, MA).
By performing substantially the same method as the flow cytometry for antibody

binding ability of Example 5, it was confirmed that both PRb-CNO1 and PRb-CN32
bispecific
antibodies scFv-C-K-scFy bound to NIH3T3 cells (FIG. 18). Specifically, the
bispecific
antibody scFv-C-K-scFy fusion protein (100 nM) in flow cytometry buffer was
cultured on
NIH3T3 cells expressing mPDGFR-f3. The cells were treated with APC-anti human
CI<
antibody. The bispecific antibody anti-HER2 x cotinine scFv-C-K-scFy fusion
protein was used
as a negative control.
It was carried out in substantially the same manner as the enzyme immunoassay
method for antibody binding ability of Example 3, except that mPDGFR-13-hFc
chimeric
protein was coated with 0/N on a microtiter plate at a temperature of 4 C.
After incubating the
blocking agent in each well, various concentrations of PRb-CN01-rabbit Fc
(0.01 nM-1 pM)
and PRb-CN01, control anti-HER2 antibody, and PRb-CN32 bispecific scFv-C-K-
scFy (100 nM)
diluted in 3% BSA/PBS were reacted in each well at 37 C for 2 hours. After
washing the plate
three times with PBST, the HRP-anti rabbit Fc antibody was reacted at 37 C.
for 1 hour, and
after washing, the reaction was performed using ABTS. Then, the absorbance was
measured at
405 nm with a Multiscan Ascent microplate device (Labsystems, Helsinki,
Finland) (FIG. 19A).
Fig. 19a is the result of confirming the competition for the PRb-CN01
surrogate antibody. As
71
Date Recue/Date Received 2021-03-31

CA 03115039 2021-03-31
shown in FIG. 19a, the binding ability of mPDGFR-13-hFc chimeric protein and
PRb-CNO1-
rabbit Fc was inhibited by PRb-CNO1 and PRb-CN32 bispecific scFv-Cx-scFv.
According to the same manner as in Example 5, PRb-CNO1-rabbit Fc (50 lily!)
and
PRb-CNO1, control antibody, PRb-CN32 bispecific antibody scFv-Cx-scFv (1 pM)
were
cultured on NIH3T3 cells for 1 hour. After washing, it was reacted with FITC-
anti rabbit Fc
antibody (172-1506, KPL, Gaithersburg, Maryland, USA) at 4 C for 1 hour. After
washing, it
was classified and analyzed with a FACS Canto II instrument (BD Biosciences,
San Jose, CA,
USA). 10,000 cells were identified for each measurement. The results were
analyzed by FlowJo
(Tree Star, Ashland, OR, USA) (Fig. 19b).
As shown in Fig. 19b, the binding ability of mPDGFR-13 and PRb-CNO1-rabbit Fc
present on the cell surface was inhibited by the PRb-CNO1 and PRb-CN32
bispecific antibody
scFv-Cx-scFv.
It was carried out in the same manner as in Example 7, except that the
bispecific
antibody related with the anti-mPDGFR-13 antibody and the bispecific antibody
related with
the anti-hPDGFR-13 antibody prepared in Example 10 were used. Fig. 20a to Fig.
20b are graphs
showing the difference in cytotoxic activity of PRb-CNO1 and PRb-CN32
bispecific antibodies
and cotinine-duocarmycin complexes in the cells expressing PDGFR-f3. In Fig.
20b, an
experiment was repeated in the presence of mPDGF-BB.
As shown in Fig. 20a and Fig. 20b, both of the two tested antibodies (PRb-
CNO1, PRb-
32) showed the equivalent cytotoxicity regardless of the presence/absence of
mPDGF-BB.
Therefore, it was confirmed that PRb-CNO1 and PRb-32 are surrogate antibodies.
72
Date Recue/Date Received 2021-03-31

Representative Drawing
A single figure which represents the drawing illustrating the invention.
Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2019-10-07
(87) PCT Publication Date 2020-04-09
(85) National Entry 2021-03-31
Examination Requested 2021-03-31

Abandonment History

There is no abandonment history.

Maintenance Fee

Last Payment of $100.00 was received on 2023-09-20


 Upcoming maintenance fee amounts

Description Date Amount
Next Payment if small entity fee 2024-10-07 $100.00
Next Payment if standard fee 2024-10-07 $277.00

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee 2021-03-31 $408.00 2021-03-31
Request for Examination 2024-10-07 $816.00 2021-03-31
Maintenance Fee - Application - New Act 2 2021-10-07 $100.00 2021-09-24
Maintenance Fee - Application - New Act 3 2022-10-07 $100.00 2022-09-20
Maintenance Fee - Application - New Act 4 2023-10-10 $100.00 2023-09-20
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
SEOUL NATIONAL UNIVERSITY R&DB FOUNDATION
EWHA UNIVERSITY-INDUSTRY COLLABORATION FOUNDATION
Past Owners on Record
None
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Abstract 2021-03-31 1 8
Claims 2021-03-31 7 217
Drawings 2021-03-31 32 1,725
Description 2021-03-31 72 3,062
Representative Drawing 2021-03-31 1 139
Patent Cooperation Treaty (PCT) 2021-03-31 1 39
Patent Cooperation Treaty (PCT) 2021-03-31 3 182
International Search Report 2021-03-31 8 404
Amendment - Abstract 2021-03-31 2 136
National Entry Request 2021-03-31 10 384
Cover Page 2021-04-27 2 171
Examiner Requisition 2022-04-01 5 299
Amendment 2022-07-27 48 2,432
Prosecution Correspondence 2022-10-17 54 3,868
Description 2022-07-27 72 4,443
Claims 2022-07-27 7 327
Drawings 2022-07-27 32 1,795
Office Letter 2023-01-06 1 215
Amendment 2023-12-21 22 950
Claims 2023-12-21 7 321
Examiner Requisition 2023-08-25 12 358

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :